EP2357184A1 - Imidazoquinoxaline compounds as immunomodulators - Google Patents

Imidazoquinoxaline compounds as immunomodulators Download PDF

Info

Publication number
EP2357184A1
EP2357184A1 EP11164995A EP11164995A EP2357184A1 EP 2357184 A1 EP2357184 A1 EP 2357184A1 EP 11164995 A EP11164995 A EP 11164995A EP 11164995 A EP11164995 A EP 11164995A EP 2357184 A1 EP2357184 A1 EP 2357184A1
Authority
EP
European Patent Office
Prior art keywords
substituted
alkyl
compound
antigens
antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP11164995A
Other languages
German (de)
French (fr)
Other versions
EP2357184B1 (en
Inventor
James Sutton
Feng Xu
Nicholas Valiante
Jiong Lan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Novartis AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis AG filed Critical Novartis AG
Publication of EP2357184A1 publication Critical patent/EP2357184A1/en
Application granted granted Critical
Publication of EP2357184B1 publication Critical patent/EP2357184B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention generally relates to small molecule immune potentiators (SMIPs) that are novel imidazoquinoxaline compounds and analogs thereof that are capable of stimulating or modulating an immune response in a subject.
  • SIPs small molecule immune potentiators
  • the invention also relates to novel combinations of antigens with the immune potentiators that may be used in vaccine therapies.
  • the compounds can be used as immunotherapeutic agents for proliferative diseases, infectious diseases, autoimmune diseases, allergies, and/or asthma.
  • U.S. Patent No. 6,083,505 describes specific imidazoquinolines for use as adjuvants.
  • WO 03/097641 discloses the use of certain imidazoquinolines and salts thereof for the treatment of certain protein kinase dependent diseases and for the manufacture of pharmaceutical preparations for the treatment of diseases.
  • Immune response to certain antigens can be enhanced through the use of immune potentiators, known as vaccine adjuvants.
  • vaccine adjuvants potentiate the immune response to specific antigens and are, therefore, the subject of considerable interest and study within the medical community.
  • vaccines possessing antigenic epitopes that were previously impossible to produce.
  • vaccine candidates include synthetic peptides mimicking numerous bacterial and viral antigens.
  • the immune response to these purified antigens can be enhanced by coadministration of an adjuvant.
  • conventional vaccine adjuvants possess a number of drawbacks that limit their overall use and effectiveness.
  • many of the adjuvants currently available have limited utility because they include components that are not metabolized by humans.
  • most adjuvants arc difficult to prepare and may require time-consuming procedures and, in some cases, the use of elaborate and expensive equipment to formulate a vaccine and adjuvant system.
  • SMIPs small molecule immune potentiators
  • Novel sole-acting agents with varied capacities for altering levels and/or profiles of cytokine production in human immune cells are needed.
  • Compounds with structural disparities will often elicit a desired response through a different mechanism of action, or with greater specificity to a target, such as a dendritic cell, modulating potency and lowering side effects when administered to a patient.
  • cytostatic substances have rendered them useful in the therapy of autoimmune diseases such as multiple sclerosis, psoriasis and certain rheumatic diseases.
  • autoimmune diseases such as multiple sclerosis, psoriasis and certain rheumatic diseases.
  • their beneficial effect has to be weighed against serious side effects that necessitate dosages that are too low.
  • interruption of the treatment may be required.
  • agents and/or combinations of active substances that result in significantly improved cytostatic or cytotoxic effects compared to conventional cytostatics are needed.
  • chemotherapies may be offered that combine increasing efficiency with a large reduction of side effects and therapeutic doses.
  • Such agents and combination therapies may thus increase the therapeutic efficiency of known cytostatic drugs.
  • the compounds of the invention are used in combination with compounds that provide significantly improved cytostatic or cytotoxic effect compared to conventional cytostatic agents when administered alone.
  • cell lines that are insensitive to conventional chemotherapeutic treatment may also be susceptible to chemotherapy using combinations of active substances.
  • the present invention provides such methods, and further provides other related advantages.
  • the current invention provides method of preparing therapeutic and prophylactic agents for treatment of disease states characterized by other immune deficiencies, abnormalities, or infections including autoimmune diseases and viral and bacterial infections responsive to compounds with the capacity to modulate cytokines and/or TNF- ⁇ .
  • the instant invention provides novel immune potentiators, immunogenic compositions, novel compounds and pharmaceutical compositions, and novel methods of administering a vaccine, by administering small molecule immune potentiators alone or in combination with antigens and/or other agents.
  • the invention further provides novel compounds and pharmaceutical compositions, for use in the treatment of cancer, precancerous lesions, autoimmune diseases, infectious diseases, allergies, and asthma.
  • the invention further provides the use of the compounds of the invention in the manufacture of medicaments for use in the treatment of cancer, precancerous lesion, autoimmune diseases, allergies, and asthma.
  • imidazoquinoxaline compounds and analogs thereof used in the methods and compositions of the invention are inexpensive to produce and easy to administer. They have potential for finer specificity compared to existing immunostimulants, thus providing improved efficacy and safety profiles.
  • the imidazoquinoxaline compounds and analogs thereof may be combined with numerous antigens and delivery systems to form an immunogenic composition.
  • the immunogenic composition can be used in the manufacture of a vaccine or a medicament.
  • the imidazoquinoxaline compounds and analogs thereof are used alone or in combination with other therapies (e.g., anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of the following: persistent of chronic viral infections such as, e.g., those caused by the human immunodeficiency virus (HIV), the hepatitis C virus (HCV), the hepatitis B virus (HBV), the herpes simplex virus (HSV); persistent or chronic bacterial infections, such as those caused by Chlamydia, pseudomonas, gonorrhea, treponema pallidium (syphilis), H.
  • HCV human immunodeficiency virus
  • HCV hepatitis C virus
  • HBV hepatitis B virus
  • HSV herpes simplex virus
  • persistent or chronic bacterial infections such as those caused by Chlamydia, pseudomonas, gonorrhea, treponema pallidium (s
  • pylori tuberculosis, Lyme disease
  • chronic or persistent fungal infections chronic or persistent parasitic infections (e.g., malaria)
  • medicaments for the reduction of tumor growth or modulation of abnormal cellular proliferation associated with diseases such as actinic kcratosis, atypical or dysplastic ncvi, or premalignant lentigos.
  • the imidazoquinoxaline compounds and analogs thereof of the present invention may target substrates in the disease state, such as, for example particular kinases including EGFr, c-Kit, bFGF, Kdr, CHK1, CDK, cdc-2, Akt, PDGF, PI3K, VEGF, PKA, PKB, src, c-Met, Abl, Ras, RAF, and MEK, among others.
  • kinases including EGFr, c-Kit, bFGF, Kdr, CHK1, CDK, cdc-2, Akt, PDGF, PI3K, VEGF, PKA, PKB, src, c-Met, Abl, Ras, RAF, and MEK, among others.
  • the imidazoquinoxaline compounds and analogs thereof may also be used for the treatment of cancer either alone or in combination with other anticancer therapies (e.g., chemotherapeutic agents, (monoclonal antibodies) mAbs or other immune potentiators).
  • chemotherapeutic agents e.g., monoclonal antibodies
  • mAbs monoclonal antibodies
  • certain imidazoquinoxalines with the capacity to induce Type 1 cytokines e.g., IL-12, TNF- ⁇ or IFN's
  • Type 1 cytokines e.g., IL-12, TNF- ⁇ or IFN's
  • the imidazoquinoxaline compounds and analogs thereof may be used, for example, for the treatment of bacillus Calmette-Guerin (BCG), cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenza b, meningococcus infection, and pneumococcus infection.
  • BCG Bacillus Calmette-Guerin
  • cholera cholera
  • plague typhoid
  • hepatitis B infection influenza
  • influenza inactivated polio
  • rabies measles
  • mumps rubella
  • oral polio yellow fever
  • tetanus diphtheria
  • hemophilus influenza b meningococcus infection
  • pneumococcus infection pneumococcus infection.
  • methods of treating cancer and/or precancerous lesions are provided.
  • one or more known anticancer agent is combined with one or more imidazoquinoxaline compound to reduce tumor growth in a subject.
  • suitable anticancer agents are contemplated for use in the methods of the present invention and are described more thoroughly in the following detailed description.
  • a method of inhibiting tumor cell growth in a subject includes administering to a subject an effective dose of a combination comprising at least one imidazoquinoxaline compound as described herein, and a monoclonal antibody (mAb).
  • the combination may be more effective at inhibiting such cell growth than when the mAb is administered by itself.
  • an additional imidazoquinoxaline compound as described herein compound and/or mAb is administered to the subject.
  • the invention provides immunogenic compositions comprising an antigen and an imidazo[1,2-a]quinoxalin-4-amine effective to stimulate a cell mediated response to said antigen.
  • the imidazo[1,2-a]quinoxalin-4-amine compounds have the general Formula described herein. Accordingly, in some embodiments of the methods and compositions of the invention, the imidazoquinoxaline compound has the Formula (I): wherein:
  • the compound has the Formula (II): wherein:
  • the compound has the Formula (II): wherein:
  • compounds of Formula (I) or (II) can be used in the manufacture of a medicament for enhancing the immune response to an antigen.
  • inventions provide the use of the compounds of the invention, in the manufacture of medicament for immune stimulation, and another agent, such as an antigen, for simultaneous separate or sequential administration.
  • the use is for treating or preventing a bacterial or viral infection.
  • the use is for treating cancer.
  • the use is for preventing influenza infection.
  • compositions comprising (a) a compound of Formula (I) or (II); and (b) an antigen, wherein (a) and (b) are either in admixture or arc separate compositions.
  • the agents arc for simultaneous separate or sequential administration.
  • the use is for preventing a viral, bacterial, fungal or parasitic infection.
  • the use is for treating cancer.
  • Applicants have discovered methods of stimulating cytokine activity in cells and immunotherapeutics and/or vaccine adjuvants, that will provide effective treatments for disorders such as those described herein and those apparent to one skilled in the art.
  • the invention provides a compound of Formula (I): wherein:
  • R 1 is not methyl
  • R 1 is not isobutyl
  • R 1 is not phenethyl
  • R 1 is C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, or substituted C 2 -C 12 alkenyl.
  • R 1 is hydrogen, C 1 -C 12 alkyl or substituted C 1 -C 12 alkyl.
  • R 1 is C 1 -C 12 alkyl.
  • R 1 is C 1 -C 6 alkyl, for example isobutyl (i.e., -CH 2 -CH(CH 3 ) 2 ).
  • R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen or hydroxyalkyl, for example -CH 2 -C(OH)(CH 3 ) 2 .
  • R 1 is C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, or substituted C 2 -C 12 alkenyl; and R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, or hydroxyalkyl.
  • R 1 is C 1 -C 12 alkyl or substituted C 1 -C 12 alkyl; and R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl or hydroxyalkyl.
  • R 1 is C 1 -C 12 alkyl; and R 2 is hydrogen, C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 1 is C 1 -C 6 alkyl; and R 2 is hydrogen or hydroxyalkyl. In some embodiments, R 1 is -CH 2 -CH(CH 3 ) 2 ; and R 2 is hydrogen or hydroxyalkyl, for example -CH 2 -C(OH)(CH 3 ) 2 .
  • R 6 is -OR 3 or -N(R 3 )(R 4 ). In other embodiments, only one of R 1 , R 2 and R 6 is H.
  • R 6 is hydrogen.
  • the compound is selected from and pharmaceutically acceptable salts, tautomers and salts of the tautomers thereof.
  • the compound is selected from the group consisting of:
  • the compound has the Formula (II): wherein:
  • R 1 is -N(R 3 )(R 4 ). More particular still, R 3 and R 4 arc each independently selected from the group consisting of hydrogen, C 1 -C 12 alkyl, and substituted C 1 -C 12 alkyl. In another embodiment R 1 is hydroxy.
  • R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen or hydroxyalkyl, for example -CH 2 -C(OH)(CH 3 ) 2 .
  • the compound has the Formula (II): wherein:
  • R a and R b are taken together to form a 6-membered substituted or unsubstituted heteroaryl group, more particularly a substituted or unsubstituted pyridyl group.
  • R a and R b are taken together to form a saturated group selected from a heterocyclyl, substituted heterocyclyl, cycloalkyl or substituted cycloalkyl.
  • R a and R b are taken together to form a substituted or unsubstituted aryl group, more particularly a substituted or unsubstituted phenyl group.
  • R 1 is C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, or substituted C 2 -C 12 alkenyl; and R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, or hydroxyalkyl.
  • R 1 is C 1 -C 12 alkyl or substituted C 1 -C 12 alkyl; and R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl or hydroxyalkyl.
  • R 1 is C 1 -C 12 alkyl; and R 2 is hydrogen, C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 1 is C 1 -C 6 alkyl; and R 2 is hydrogen or hydroxyalkyl. In some embodiments, R 1 is -CH 2 -CH(CH 3 ) 2 ; and R 2 is hydrogen or hydroxyalkyl, for example -CH 2 -C(OH)(CH 3 ) 2 .
  • R 1 is -N(R 3 )(R 4 ). More particular still, R 3 and R 4 are each independently selected from the group consisting of hydrogen, C 1 -C 12 alkyl, and substituted C 1 -C 12 alkyl. In another embodiment R 1 is hydroxy.
  • R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen, C 1 -C 12 alkyl or hydroxyalkyl. In some embodiments, R 2 is hydrogen or hydroxyalkyl, for example -CH 2 -C(OH)(CH 3 ) 2 .
  • R a and R b together form an unsubstituted phenyl group; R 1 and R 2 are not simultaneously hydrogen, and if R 2 is hydrogen, then R 1 is not methyl, isobutyl, or phenethyl.
  • any asymmetric carbon atom(s) can have either the R or S configuration.
  • Substituents at a double bond or a ring of the compounds of Formula (I) or (II) may be present in either the cis (-Z-) or trans (-E-) configurations.
  • the compounds may thus be present as mixtures of isomers, diastereomers, and enantiomers or may be present as pure isomers.
  • the compounds are enantiomerically pure where only one enantiomer is present.
  • the compound may be present as a mixture of enantiomers which includes more of one enantiomer than it does of the other.
  • a compound of the invention is considered effective to elicit an immune response at a concentration of 300 ⁇ M or less in some embodiments, 200 ⁇ M or less in some embodiments, 100 ⁇ M or less in some embodiments, or 20 ⁇ M or less in some embodiments if the compound of the invention effects (a) the production of TNF- ⁇ in an in vitro cell based assay of human peripheral blood mononuclear cells, and (b) a concentration of human peripheral blood mononuclear cells (PBMCs) of about 500,000/mL, when the cells are exposed to the compound for a period of about 18-24 hours, preferably about 24 hours.
  • PBMCs human peripheral blood mononuclear cells
  • the above method of stimulating a local immune response includes the stimulation of a local immune response where the selected cells or tissues are infected or cancerous.
  • the selected cells or tissues are infected with a fungus or bacterium.
  • the selected tissues are inflamed with an allergen, for example in an asthmatic condition.
  • the selected cells are infected with a virus or bacteria.
  • the infectious agent is HCV, HIV, HBV, HSV, H. pylori, HSV Type 1 or 2, or Human Papilloma Virus.
  • Another embodiment provides a method of inducing interferon biosynthesis in a subject.
  • Such methods include administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce interferon biosynthesis.
  • a vaccine adjuvant of Formula (I) or (II) is administered to the subject in an amount sufficient to induce interferon biosynthesis.
  • Another embodiment provides a compound of Formula (I) or (II), wherein the compound is co-administered with another agent to a patient in need thereof.
  • the agent is an antigen or a vaccine.
  • the compound of Formula (I) or (II) may be administered to the subject before, during, or after the other agent is administered to the subj ect. Therefore, in some embodiments, the compound of Formula (I) or (II) is administered to the subject at the same time that the other agent is administered to the subject.
  • the location or site of administration of the compound of Formula (I) or (II) can be the same or different as the location of an antigen when the compound is used with an antigen.
  • Another embodiment provides a method of modulating an immune response in a subject. Such methods include administering a compound of Formula (I) or (II) to the subject.
  • Another embodiment provides a method for inducing the production of TNF- ⁇ in a subject.
  • Such methods include administering a compound of Formula (I) or (II) to a subject in an amount sufficient to induce the production of TNF- ⁇ .
  • the compound has an average steady state drug concentration in the blood of less than 20 ⁇ M.
  • Another embodiment provides a method of inducing an immune response in a subject.
  • the embodiment includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • the immune response includes the production of cytokines or increased production of TNF- ⁇ .
  • the induction of an immune response includes the production of antibodies that may be neutralizing antibodies or antibodies that mediate Antibody Dependent Cell Mediated Cytotoxicity (ADCC antibodies).
  • Another embodiment provides a method of inducing an immune response in a subject suffering from a microbial (viral, bacterial, fungal or parasitic) infection.
  • the method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from a viral infection or a disease condition caused by a virus.
  • the method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response in the subject.
  • the virus may be selected from one or more of the viral pathogens described in the antigen section below.
  • the subject is suffering from a viral infection or disease condition caused by the hepatitis C virus (HCV).
  • HCV hepatitis C virus
  • the subject is suffering from a viral infection or disease condition caused by the human immunodeficiency virus (HIV).
  • the immune response is induced un a subject suffering from a bacterial, fungal or parasitic infection wherein the disease causing organism may be selected from one or more of the bacterial, fungal or parasitic pathogens described in the antigen section below.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from an abnormal cellular proliferation or cancer.
  • the method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • the compound is administered to a subject that is suffering from a disease associated with abnormal cellular proliferation.
  • the disease is selected from neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • inventions provide methods of inducing an immune response in a subject suffering from an allergic disease. Such methods include administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from asthma.
  • the method includes administering a compound of Formula (I) or (II) to the subj ect in an amount sufficient to induce an immune response.
  • asthma may be treated by steering the immune response away from Type 2 cytokine secretion and effector mechanism (e.g., IgE production and/or mast cell/basophil activation).
  • Another embodiment provides a method of inducing an immune response in a subject suffering from precancerous lesions.
  • the method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • the precancerous lesions are actinic keratosis.
  • the precancerous lesions are selected from actinic keratosis, atypical or dysplastic nevi, or premalignant lentigos.
  • the compound of Formula (I) or (II) is administered topically to a subject.
  • inventions provide a method of inhibiting a kinase in a subject. Such methods include administering the compound of Formula (I) or (II) to the subject.
  • Another embodiment provides a method of modulating an immune response in a subject.
  • the method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to inhibit a kinase in the subject.
  • the kinase is selected from EGFr, c-Kit, bFGF, Kdr, CHK1, CDK, cdc-2, Akt, PDGF, PI3K, VEGF, PKA, PKB, src, c-Met, Abl, Ras, RAF, MEK, or combinations thereof.
  • the compound of Formula (I) is administered topically to a subject.
  • Another embodiment provides a method of inducing an immune response in a subject, comprising: administering to the subject a compound of Formula (I) or (II) and an antigen, wherein the compound induces or enhances an immune response to the antigen in the subject.
  • the antigen can be one or more viral, bacterial, fungal, parasitic or tumor antigens or other antigens as described herein.
  • compositions comprising: the compound of Formula (I) or (II) and another agent.
  • the other agent is an antigen.
  • the composition of the invention comprises the compound of Formula (I) or (II) with an antigen and a second adjuvant.
  • the composition of the invention provides a compound of Formula (I) or (II) and a second adjuvant.
  • the composition further comprises poly(lactide-co-glycolide) (PLG).
  • PLG poly(lactide-co-glycolide)
  • the composition further comprises MF59 or another adjuvant.
  • Another embodiment provides a pharmaceutical composition, comprising: the compound of Formula (I) or (II) and a pharmaceutically acceptable excipient.
  • Another embodiment of the present invention provides a method of stimulating TLR-7 production comprising administering a compound of Formula (I), or (II)
  • Another embodiment provides a method of stimulating TLR-8 production comprising administering a compound of Formula (I).
  • Another embodiment provides a method of stimulating TLR-7 and TLR-8 production comprising administering a compound of Formula (I) or (II).
  • Compounds of the present invention cause immune potentiation and stimulate production of TLR-7 and TLR-8. Such compounds can be used as polyclonal activators for the production of antigens. More particularly the invention relates to a method of preparing monoclonal antibodies with a desired antigen specificity comprising contacting the compounds of the present invention (such as those of Formula (I) or (II)) with immortalized memory B cells.
  • the monoclonal antibodies produced therefrom, or fragments thereof may be used for the treatment of disease, for the prevention of disease or for the diagnosis of disease.
  • Methods of diagnosis may include contacting an antibody or an antibody fragment with a sample.
  • the methods of diagnosis may also include the detection of an antigen/antibody complex.
  • the memory B cells to be transformed can come from various sources (e.g. from whole blood, from peripheral blood mononuclear cells (PBMCs), from blood culture, from bone marrow, from organs, etc.), and suitable methods for obtaining human B cells are well known in the art. Samples may include cells that are not memory B cells or other blood cells. A specific human memory B lymphocyte subpopulation exhibiting a desired antigen specificity may be selected before the transformation step by using methods known in the art. In one embodiment, the human memory B lymphocyte subpopulation has specificity for a virus e.g. the B cells are taken from a patient who is suffering or has recovered from the virus. In another embodiment, B cells are taken from subjects with Alzheimer's disease and include B cells with specificity for B-amyloid (e.g. Mattson & Chan (2003) Science 301:1 847-9 ; etc.).
  • B-amyloid e.g. Mattson & Chan (2003) Science 301:1 847-9 ; etc.
  • Another embodiment provides a method for producing immortalized B memory lymphocytes, comprising the step of transforming B memory lymphocytes using the Epstein Barr virus in the presence of a compound of the present invention, such as a compound of Formula (I) or (II). See WO 04/76677 .
  • compositions that include any of the aforementioned compounds or embodiments of Formula (I) or (II).
  • Such compositions may include other pharmaceutically acceptable ingredients such as one or more of excipients, carriers, and the like well-known to those skilled in the art.
  • R 6 of the compounds of Formula (I) is hydrogen.
  • the imidazoquinoxaline compounds and analogs thereof can be used with or without an antigen in therapeutic applications, for example to treat cancer or infectious diseases.
  • the imidazoxaquinoline compounds may also be used in combination with other therapeutic agents, such as anti-viral agents and monoclonal antibodies in different therapeutic applications.
  • One embodiment of the method of inducing an immunostimulatory effect in a patient is directed to administering an immunogenic composition comprising an antigen in an amount effective to stimulate an immune response such as a cell-mediated immune response and, as a vaccine adjuvant, an imidazoquinoline compound, in an amount effective to potentiate the immune response such as the cell-mediated immune response to the antigen.
  • Agents combined with the imidazoquinoxaline compounds and analogs thereof, contemplated to be useful in treating the aforementioned diseases include those well known in the art, such as, but not limited to, anesthetics, hypnotic sedatives, anti-anxieties, antiepileptics, antipyretic antiphlogistics, stimulants, wake amines, anti-Parkinson drugs, agents for psychoneuroses, agents for central nervous system, skeletal muscle relaxants, agents for autonomic nervous system, antispastic agents, cytotoxic agents, monoclonal antibodies, drugs for eye, drugs for nose and ear, anti-vertiginous drugs, cardiotonics, antiarrhythmic drugs, diuretics, pressure reduction drugs, vasoconstrictors, coronary vasodilators, peripheral vasodilating drugs, hyper-lipemia drugs, breath stimulants, antitussive and expectorant drugs, bronchodilators, drugs for allergy, antidiarrheal drugs, drugs for intestinal disorders,
  • compositions described herein are used for the treatment of cancer and reduction of tumor growth.
  • an imidazoxaquinoline compound of the invention is combined with a known mAb for the treatment of cancer.
  • an antibody and an imidazoxaquinoline compound are administered to a subject in need thereof.
  • the antibody individually, has an inhibiting effect upon tumor cell growth, and the imidazoxaquinoline compound induces the production of cytokines.
  • the combination is expected to be more effective at inhibiting the growth of certain mammalian tumor cells than are any of the agents when individually administered.
  • methods of treating cancer are provided in which known anticancer agents are combined with imidazoquinoxaline compounds and analogs thereof of the invention to reduce tumor growth in a subject.
  • suitable anticancer agents arc contemplated for use in such methods.
  • the present invention contemplates, but is not limited to, administration of numerous anticancer agents including, but not limited to: fcnrctinidc, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortczomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib (iressa), erlotinib (tarceva), perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, staurosporine, alvocidib, genistein, DA-9601, avi
  • anticancer agents comprise agents that induce or stimulate apoptosis.
  • Agents that induce apoptosis include, but are not limited to, radiation (e.g., W); kinase inhibitors (e.g., Epidermal Growth Factor Receptor [EGFR] kinase; inhibitor, Vascular Growth Factor Receptor [VGFR] kinase inhibitor, Fibroblast Growth 5 Factor Receptor [FGFR] kinase inhibitor, Platelet-derived Growth Factor Receptor [PGFR] I kinase inhibitor, EGFr and Bcr-Ab1 kinase inhibitors such as Gleevec, Iressa, and Tarceva]); antisense molecules; antibodies [e.g., Herceptin and Rituxan]; anti-estrogens [e.g., raloxifene and tamoxifen]; anti-androgens [e.g., flutamide, bicalutamide, finasteride
  • Anti-inflammatory drugs I NSAIDs
  • cancer chemotherapeutic drugs e.g., CPT-11, fludarabine (Fludara), dacarbazine (DTIC), dexamethasone, mitoxantrone, Mylotarg, cisplatinum, 5-FU, Doxrubicin, Taxotere or taxol
  • cellular signaling molecules ceramides and cytokines; and the like may also be administered to subjects in conjunction with the imidazoquinoxalines of Formula (I) or (II).
  • methods of treating allergies include administering an imidazoquinoxaline compound alone or in combination with another agent known to be effective against allergies.
  • the combination is more effective in treating an allergic condition than the known agcnt(s) is/are without the addition of the imidazoquinoxaline compound.
  • the known agent is an antihistamine and/or a leukotriene inhibitor.
  • the allergic condition is asthma.
  • the allergic condition is selected from allergic rhinitis, dermatosis, or urticaria.
  • the combination is administered to the subject enterally, parenterally, intranasally, subcutaneously, or intraarterially.
  • compositions contemplated to be within the scope of the present invention may include (an) additional adjuvant(s) and or other immune stimulator compound.
  • compositions may be administered in conjunction with other immunoregulatory agents.
  • compositions can include an adjuvant.
  • adjuvants for use with the invention include, but are not limited to, one or more of the following set forth below:
  • Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminum salts and calcium salts.
  • the invention includes mineral salts such as hydroxides ( e.g . oxyhydroxides), phosphates ( e.g . hydroxyphosphates, orthophosphates), sulfates, etc. ( e.g. see chapters 8 & 9 of Vaccine Design... (1995) eds. Powell & Newman. ISBN: 030644867X. Plenum .), or mixtures of different mineral compounds ( e.g . a mixture of a phosphate and a hydroxide adjuvant, optionally with an excess of the phosphate), with the compounds taking any suitable form ( e.g . gel, crystalline, amorphous, etc. ), and with adsorption to the salt(s) being preferred.
  • the mineral containing compositions may also be formulated as a particle of metal salt ( WO00/23105 ).
  • Aluminum salts may be included in vaccines of the invention such that the dose of Al 3+ is between 0.2 and 1.0 mg per dose.
  • the aluminum based adjuvant for use in the present invention is alum (aluminum potassium sulfate (AlK(SO 4 ) 2 )), or an alum derivative, such as that formed in-situ by mixing an antigen in phosphate buffer with alum, followed by titration and precipitation with a base such as ammonium hydroxide or sodium hydroxide.
  • alum aluminum potassium sulfate (AlK(SO 4 ) 2 )
  • AlK(SO 4 ) 2 aluminum potassium sulfate
  • Aluminum-based adjuvant for use in vaccine formulations of the present invention is aluminum hydroxide adjuvant (Al(OH) 3 ) or crystalline aluminum oxyhydroxide (AlOOH), which is an excellent adsorbant, having a surface area of approximately 500m 2 /g.
  • Al(OH) 3 aluminum hydroxide adjuvant
  • AlOOH crystalline aluminum oxyhydroxide
  • AlPO 4 aluminum phosphate adjuvant
  • AlPO 4 aluminum hydroxyphosphate, which contains phosphate groups in place of some or all of the hydroxy groups of aluminum hydroxide adjuvant is provided.
  • Preferred aluminum phosphate adjuvants provided herein are amorphous and soluble in acidic, basic and neutral media.
  • the adjuvant of the invention comprises both aluminum phosphate and aluminum hydroxide.
  • the adjuvant has a greater amount of aluminum phosphate than aluminum hydroxide, such as a ratio of 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1 or greater than 9:1, by weight aluminum phosphate to aluminum hydroxide.
  • aluminum salts in the vaccine are present at 0.4 to 1.0 mg per vaccine dose, or 0.4 to 0.8 mg per vaccine dose, or 0.5 to 0.7 mg per vaccine dose, or about 0.6 mg per vaccine dose.
  • the preferred aluminum-based adjuvant(s), or ratio of multiple aluminum-based adjuvants, such as aluminum phosphate to aluminum hydroxide is selected by optimization of electrostatic attraction between molecules such that the antigen carries an opposite charge as the adjuvant at the desired pH.
  • adsorbs lysozyme but not albumin at pH 7.4.
  • albumin be the target
  • aluminum hydroxide adjuvant would be selected (iep 11.4).
  • pretreatment of aluminum hydroxide with phosphate lowers its isoelectric point, making it a preferred adjuvant for more basic antigens.
  • Oil-emulsion compositions suitable for use as adjuvants in the invention include squalene-water emulsions, such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer). See WO90/14837 .
  • Particularly preferred adjuvants for use in the compositions are submicron oil-in-water emulsions.
  • Preferred submicron oil-in-water emulsions for use herein are squalene/water emulsions optionally containing varying amounts of MTP-PE, such as a submicron oil-in-water emulsion containing 4-5% w/v squalene, 0.25-1.0% w/v Tween 80TM (polyoxyelthylenesorbitan monooleate), and/or 0.25-1.0% Span 85TM (sorbitan trioleate), and, optionally, N-acetylmuramyl-L-alanyl-D-isogluatminyl-L-alanine-2-(1'-2'-dipalmitoylsn-glycero-3-huydroxyphosphophoryloxy)-ethylamine (MTP-PE), for example, the submicron oil-in-water emul
  • MF59 -- Design and Evaluation of a Safe and Potent Adjuvant for Human Vaccines in Vaccine Design: The Subunit and Adjuvant Approach (Powell, M.F. and Newman, M.J. eds.) Plenum Press, New York, 1995, pp. 277-296 ).
  • MF59 contains 4-5% w/v Squalene (e.g.
  • MTP-PE may be present in an amount of about 0-500 ⁇ g/dose, more preferably 0-250 ⁇ g/dose and most preferably, 0-100 ⁇ g/dose.
  • MF59-0 refers to the above submicron oil-in-water emulsion lacking MTP-PE, while the term MF59-MTP denotes a formulation that contains MTP-PE.
  • MF59-100 contains 100 ⁇ g MTP-PE per dose, and so on.
  • MF69 another submicron oil-in-water emulsion for use herein, contains 4.3% w/v squalene, 0.25% w/v Tween 80TM, and 0.75% w/v Span 85TM and optionally MTP-PE.
  • MF75 also known as SAF, containing 10% squalene, 0.4% Tween 80TM, 5% pluronic-blocked polymer L121, and thr-MDP, also microfluidized into a submicron emulsion.
  • MF75-MTP denotes an MF75 formulation that includes MTP, such as from 100-400 ⁇ g MTP-PE per dose.
  • Submicron oil-in-water emulsions, methods of making the same and immunostimulating agents, such as muramyl peptides, for use in the compositions, arc described in detail in International Publication No. WO90/14837 and US Patent Nos. 6,299,884 and 6,45 1,325 .
  • Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IFA) may also be used as adjuvants in the invention.
  • oil-in-water emulsion adjuvants useful with the invention include, but are not limited to:
  • the emulsions are preferably mixed with additional agents (such as an antigen) extemporaneously, at the time of delivery.
  • additional agents such as an antigen
  • the adjuvant and antigen are typically kept separately in a packaged or distributed vaccine, ready for final formulation at the time of use.
  • the antigen will generally be in an aqueous form, such that the vaccine is finally prepared by mixing two liquids.
  • the volume ratio of the two liquids for mixing can vary ( e.g . between 5:1 and 1:5) but is generally about 1:1.
  • composition includes a tocopherol
  • any of the ⁇ , ⁇ , ⁇ , ⁇ , ⁇ or ⁇ tocopherols can be used, but ⁇ -tocopherols are preferred.
  • the tocopherol can take several forms e.g . different salts and/or isomers. Salts include organic salts, such as succinate, acetate, nicotinate, etc. D- ⁇ -tocopherol and DL- ⁇ -tocopherol can both be used.
  • Tocopherols are advantageously included in vaccines for use in elderly patients ( e.g . aged 60 years or older) because vitamin E has been reported to have a positive effect on the immune response in this patient group [ Han et al.
  • a preferred ⁇ -tocopherol is DL- ⁇ -tocopherol, and the preferred salt of this tocopherol is the succinate.
  • the succinate salt has been found to cooperate with TNF-related ligands in vivo.
  • ⁇ -tocopherol succinate is known to be compatible with influenza vaccines and to be a useful preservative as an alternative to mercurial compounds
  • Saponin formulations may also be used as adjuvants in the invention.
  • Saponins are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponins isolated from the bark of the Quillaia saponaria Molina tree have been widely studied as adjuvants. Saponins can also be commercially obtained from Smilax ornata (sarsaprilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root).
  • Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
  • Saponin compositions have been purified using High Performance Thin Layer Chromatography (HP-TLC) and Reversed Phase High Performance Liquid Chromatography (RP-HPLC). Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C.
  • the saponin is QS21.
  • a method of production of QS21 is disclosed in US Patent No. 5,057,540 .
  • Saponin formulations may also comprise a sterol, such as cholesterol (see WO96/33739 ).
  • ISCOMs Immunostimulating Complexes
  • cholcstcrols can be used to form unique particles called Immunostimulating Complexes (ISCOMs).
  • ISCOMs typically also include a phospholipid such as phosphatidylethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs.
  • the ISCOM includes one or more of Quil A, QHA and QHC.
  • ISCOMS may be devoid of (an) additional detergent(s). See WO00/07621 .
  • VLPs Virosomes and Virus Like Particles
  • Virosomes and Virus Like Particles can also be used as adjuvants in the invention.
  • These structures generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally non-pathogenic, nonreplicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses.
  • viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA), Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and-Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA-phages, Q ⁇ -phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein p1).
  • influenza virus such as HA or NA
  • Hepatitis B virus such as core or capsid proteins
  • Hepatitis E virus measles virus
  • Sindbis virus Rotavirus
  • Foot-and-Mouth Disease virus Retrovirus
  • Norwalk virus Norwalk virus
  • human Papilloma virus HIV
  • RNA-phages Q ⁇ -phage (such as coat proteins)
  • GA-phage such as fr-phage
  • VLPs are discussed further in WO03/024480 , WO03/024481 , and Niikura et al., "Chimeric Recombinant Hepatitis E Virus-Like Particles as an Oral Vaccine Vehicle Presenting Foreign Epitopes", Virology (2002) 293:273-280 ; Lenz et al., “Papillomarivurs-Like Particles Induce Acute Activation of Dendritic Cells", Journal of Immunology (2001) 5246-5355 ; Pinto, et al., “Cellular Immune Responses to Human Papillomavirus (HPV)-16 L1 Healthy Volunteers Immunized with Recombinant HPV-16 L1 Virus-Like Particles", Journal of Infectious Diseases (2003) 188:327-338 ; and Gerber et al., "Human Papillomavirus Virus-Like Particles Are Efficient Oral Immunogens when Coadministere
  • Virosomes are discussed further in, for example, Gluck et al., "New Technology Platforms in the Development of Vaccines for the Future", Vaccine (2002) 20:B10 -B16 .
  • Immunopotentiating reconstituted influenza virosomes are used as the subunit antigen delivery system in the intranasal trivalent INFLEXALTM product ⁇ Mischler & Metcalfe (2002) Vaccine 20 Suppl 5:B17-23 ⁇ and the INFLUVAC PLUSTM product.
  • Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as:
  • Such derivatives include Monophosphoryl lipid A (MPL) and 3-O-deacylated MPL (3dMPL).
  • 3dMPL is a mixture of 3 De-O-acylated monophosphoryl lipid A with 4, 5 or 6 acylated chains.
  • a preferred "small particle" form of 3 De-O-acylated monophosphoryl lipid A is disclosed in EP 0 689 454 .
  • Such "small particles" of 3dMPL are small enough to be sterile filtered through a 0.22 micron membrane (see EP 0 689 454 ).
  • Other non-toxic LPS derivatives include monophosphoryl lipid A mimics, such as aminoalkyl glucosaminide phosphate derivatives e.g . RC-529. See Johnson et al. (1999) Bioorg Med Chem Lett 9:2273-2278 .
  • 3dMPL has been prepared from a heptoseless mutant of Salmonella minnesota. It activates cells of the monocyte/macrophage lineage and stimulates release of several cytokines, including IL-1, IL-12, TNF- ⁇ and GM-CSF (see also ref. Thompson et al. (2005) J Leukoc Biol 78: 'The low-toxicity versions of LPS, MPL® adjuvant and RC529, are efficient adjuvants for CD4+ T cells' .). Preparation of 3dMPL was originally described in reference UK patent application GB-A-2220211 .
  • 3dMPL can take the form of a mixture of related molecules, varying by their acylation (e.g . having 3, 4, 5 or 6 acyl chains, which may be of different lengths).
  • the two glucosamine (also known as 2-deoxy-2-amino-glucose) monosaccharides are N-acylated at their 2-position carbons ( i.e . at positions 2 and 2'), and there is also O-acylation at the 3' position.
  • the group attached to carbon 2 has formula -NH-CO-CH 2 -CR 1 R 1' .
  • the group attached to carbon 2' has formula -NH-CO-CH 2 -CR 2 R 2' .
  • the group attached to carbon 3' has formula -O-CO-CH 2 -CR 3 R 3' .
  • a representative structure is:
  • Groups R 1 , R 2 and R 3 are each independently -(CH 2 ) n -CH 3 .
  • the value of n is preferably between 8 and 16, more preferably between 9 and 12, and is most preferably 10.
  • Groups R 1' , R 2' and R 3' can each independently be: (a) -H; (b) hydroxy; or (c) - O-CO-R 4 ,where R 4 is either -H or -(CH 2 ) m -CH 3 , wherein the value of m is preferably between 8 and 16, and is more preferably 10, 12 or 14. At the 2 position, m is preferably 14. At the 2' position, m is preferably 10. At the 3' position, m is preferably 12.
  • Groups R 1' , R 2' and R 3' are thus preferably -O-acyl groups from dodecanoic acid, tetradecanoic acid or hexadecanoic acid.
  • the 3dMPL has only 3 acyl chains (one on each of positions 2, 2' and 3').
  • the 3dMPL can have 4 acyl chains.
  • the 3dMPL can have 5 acyl chains.
  • the 3dMPL can have 6 acyl chains.
  • the 3dMPL adjuvant used according to the invention can be a mixture of these forms, with from 3 to 6 acyl chains, but it is preferred to include 3dMPL with 6 acyl chains in the mixture, and in particular to ensure that the hexaacyl chain form makes up at least 10% by weight of the total 3dMPL e.g . ⁇ 20%, ⁇ 30%, ⁇ 40%, ⁇ 50% or more. 3dMPL with 6 acyl chains has been found to be the most adjuvant-active form.
  • 3dMPL for inclusion in compositions of the invention is:
  • references to amounts or concentrations of 3dMPL in compositions of the invention refer to the combined 3dMPL species in the mixture.
  • 3dMPL can form micellar aggregates or particles with different sizes e.g . with a diameter ⁇ 150 nm or >500 nm. Either or both of these can be used with the invention, and the better particles can be selected by routine assay. Smaller particles (e.g . small enough to give a clear aqueous suspension of 3dMPL) are preferred for use according to the invention because of their superior activity [ WO 94/21292 ]. Preferred particles have a mean diameter less than 220 nm, more preferably less than 200 nm or less than 150 nm or less than 120 nm, and can even have a mean diameter less than 100 nm.
  • the mean diameter will not be lower than 50 nm. These particles are small enough to be suitable for filter sterilization. Particle diameter can be assessed by the routine technique of dynamic light scattering, which reveals a mean particle diameter. Where a particle is said to have a diameter of x nm, there will generally be a distribution of particles about this mean, but at least 50% by number ( e.g . ⁇ 60%, ⁇ 70%, ⁇ 80%, ⁇ 90%, or more) of the particles will have a diameter within the range x ⁇ 25%.
  • 3dMPL can advantageously be used in combination with an oil-in-water emulsion. Substantially all of the 3dMPL may be located in the aqueous phase of the emulsion.
  • the 3dMPL can be used on its own, or in combination with one or more further compounds.
  • 3dMPL in combination with the QS21 saponin [ WO94/00153 .] (including in an oil-in-water emulsion [ WO95/17210 ]), with an immunostimulatory oligonucleotide, with both QS21 and an immunostimulatory oligonucleotide, with aluminum phosphate [ WO96/26741 ], with aluminum hydroxide [ WO93/19780 ], or with both aluminum phosphate and aluminum hydroxide.
  • Lipid A derivatives include derivatives of lipid A from Escherichia coli such as OM-174.
  • OM-174 is described for example in Meraldi et al., "OM-174, a New Adjuvant with a Potential for Human Use, Induces a Protective Response with Administered with the Synthetic C-Terminal Fragment 242-310 from the circumsporozoite protein of Plasmodium berghei", Vaccine (2003) 21:2485-2491 ; and Pajak, et al., "The Adjuvant OM-174 induces both the migration and maturation of murine dendritic cells in vivo", Vaccine (2003) 21:836-842 .
  • Immunostimulatory oligonucleotides suitable for use as adjuvants in the invention include nucleotide sequences containing a CpG motif (a sequence containing an unmethylated cytosine followed by guanosine and linked by a phosphate bond). Bacterial double stranded RNA or oligonucleotides containing palindromic or poly(dG) sequences have also been shown to be immunostimulatory.
  • the CpG's can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or single-stranded.
  • the guanosine may be replaced with an analog such as 2'-deoxy-7-deazaguanosine. See Kandimalla, et al., "Divergent synthetic nucleotide motif recognition pattern: design and development of potent immunomodulatory oligodeoxyribonucleotide agents with distinct cytokine induction profiles", Nucleic Acids Research (2003) 31(9): 2393-2400 ; WO02/26757 and WO99/62923 for examples of possible analog substitutions.
  • CpG oligonucleotides The adjuvant effect of CpG oligonucleotides is further discussed in Krieg, "CpG motifs: the active ingredient in bacterial extracts?", Nature Medicine (2003) 9(7): 831-835 ; McCluskie, et al., "Parenteral and mucosal prime-boost immunization strategies in mice with hepatitis B surface antigen and CpG DNA", FEMS Immunology and Medical Microbiology (2002) 32:179-185 ; WO98/40100 ; US Patent No. 6,207,646 ; US Patent No. 6,239,116 and US Patent No. 6,429,199 .
  • the CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT. See Kandimalla, et al., "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic CpG DNAs", Biochemical Society Transactions (2003) 31 (part 3): 654-658 .
  • the CpG sequence may be specific for inducing a Th1 immune response, such as a CpG-A ODN, or it may be more specific for inducing a B cell response, such a CpG-B ODN.
  • CpG-A and CpG-B ODNs are discussed in Blackwell, et al., "CpG-A-Induced Monocyte IFN-gamma-Inducible Protein-10 Production is Regulated by Plasmacytoid Dendritic Cell Derived IFN-alpha", J. Immunol. (2003) 170(8):4061-4068 ; Krieg, “From A to Z on CpG”, TRENDS in Immunology (2002) 23(2): 64-65 and WO01/95935 .
  • the CpG is a CpG-A ODN.
  • CpG nucleotides include the following sequences, which may contain phosphorothioate modified internucleotide linkages: See WO 05/25614 .
  • the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition.
  • two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers".
  • Kandimalla "Secondary structures in CpG oligonucleotides affect immunostimulatory activity" BBRC (2003) 306:948-953 ; Kandimalla, et al., "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic GpG DNAs", Biochemical Society Transactions (2003) 31(part 3):664-658 ; Bhagat et al., "CpG penta- and hexadeoxyribonucleotides as potent immunomodulatory agents” BBRC (2003) 300:853-861 and WO03/035836 .
  • ADP-ribosylating toxins and detoxified derivatives thereof.
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention.
  • the protein is derived from E. coli (i.e., E. coli heat labile enterotoxin "LT), cholera ("CT"), or pertussis ("PT").
  • LT E. coli heat labile enterotoxin
  • CT cholera
  • PT pertussis
  • the use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in WO95/17211 and as parenteral adjuvants in WO98/42375 .
  • the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LTR192G.
  • ADP-ribosylating toxins and detoxified derivatives thereof, particularly LT-K63 and LT-R72, as adjuvants can be found in the following references: Beignon, et al., "The LTR72 Mutant of Heat-Labile Enterotoxin of Escherichia coli Enhances the Ability of Peptide Antigens to Elicit CD4+ T Cells and Secrete Gamma Interferon after Coapplication onto Bare Skin", Infection and Immunity (2002) 70(6):3012-3019 ; Pizza, et al., "Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants", Vaccine (2001) 19:2534-2541 ; Pizza, et al., "LTK63 and LTR72, two mucosal adjuvants ready for clinical trials" Int.
  • Numerical reference for amino acid substitutions is preferably based on the alignments of the A and B subunits of ADP-ribosylating toxins set forth in Domenighini et al., Mol. Microbiol (1995) 15(6):1165-1167 . Bioadhesives and Mucoadhesives
  • Bioadhesives and mucoadhesives may also be used as adjuvants in the invention.
  • Suitable bioadhesives include esterified hyaluronic acid microspheres ( Singh et al. (2001) J. Cont. Rele. 70:267-276 ) or mucoadhesives such as cross-linked derivatives of polyacrylic acid, polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides and carboxymethylcellulose. Chitosan and derivatives thereof may also be used as adjuvants in the invention. E.g. WO99/27960 .
  • Microparticles may also be used as adjuvants in the invention.
  • Microparticles i.e . a particle of ⁇ 100nm to ⁇ 150 ⁇ m in diameter, more preferably ⁇ 200nm to ⁇ 30 ⁇ m in diameter, and most preferably ⁇ 500nm to ⁇ 10 ⁇ m in diameter
  • materials that are biodegradable and non-toxic e.g . a poly( ⁇ -hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc .
  • a negatively-charged surface e.g . with SDS
  • a positively-charged surface e.g . with a cationic detergent, such as CTAB
  • liposome formulations suitable for use as adjuvants are described in US Patent No. 6,090,406 , US Patent No. 5,916,588 , and EP 0 626 169 .
  • Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters. WO99/52549 . Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol ( WO01/21207 ) as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol ( WO01/21152 ).
  • Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether (laureth 9), polyoxyethylene-9-steoryl ether, polyoxytheylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
  • PCPP Polyphosphazene
  • PCPP formulations are described, for example, in Andrianov et al., "Preparation of hydrogel microspheres by coacervation of aqueous polyphophazene solutions", Biomaterials (1998) 19(1-3):109-115 and Payne et al., “Protein Release from Polyphosphazene Matrices", Adv. Drug. Delivery Review (1998) 31(3):185-196 .
  • muramyl peptides suitable for use as adjuvants in the invention include N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-1-alanyl-d-isoglutamine (nor-MDP), and N-acetylmuramyl-1-alanyl-d-isoglutaminyl-1-alanine-2-(1'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-ethylamine MTP-PE).
  • thr-MDP N-acetyl-muramyl-L-threonyl-D-isoglutamine
  • nor-MDP N-acetyl-normuramyl-1-alanyl-d-isoglutamine
  • SIPs Small Molecule Immunopontentiators
  • imidazoquinoline compounds suitable for use adjuvants in the invention include Imiquimod and its analogues, described further in Stanley, “Imiquimod and the imidazoquinolines: mechanism of action and therapeutic potential” Clin Exp Dermatol (2002) 27(7):571-577 ; Jones, “Resiquimod 3M", Curr Opin Investig Drugs (2003) 4(2):214-218 ; Wu et al. (2004) Antiviral Res. 64(2):79-83 Vasilakos et al. (2000) Cell Immunol.
  • Preferred SMIPs include:
  • a nucleoside analog such as: (a) Isatorabine (ANA-245; 7-thia-8-oxoguanosine): and prodrugs thereof; (b)ANA975; (c) ANA-025-1; (d) ANA380; (e) the compounds disclosed in references US 6,924,271 to US2005/0070556 US 5,658,731 ; (f) a compound having the Formula: wherein:
  • thiosemicarbazone compounds as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in WO04/60308 .
  • the thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclcar cells for the production of cytokines, such as TNF- ⁇ .
  • tryptanthrin compounds as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in WO04/64759 .
  • the tryptanthrin compounds are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF- ⁇ .
  • Human immunomodulators suitable for use as adjuvants in the invention include cytokines, such as interleukins (e.g . IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc .), interferons (e.g . interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • cytokines such as interleukins (e.g . IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc .), interferons (e.g . interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • Aluminum salts and MF59 are preferred adjuvants for use with injectable i vaccines.
  • Bacterial toxins and bioadhesives are preferred adjuvants for use with mucosally-delivered vaccines, such as nasal vaccines.
  • TLR agonist it is meant a component which is capable of causing a signalling response through a TLR signalling pathway, either as a direct ligand or indirectly through generation of endogenous or exogenous ligand ( Sabroe et al, J1 2003 p1630-5 ).
  • TLR agonists of the present invention include agonists of the following:
  • TLR modulators are agonists of TLR7 (e.g. imidazoquinolines) and/or TLR9 (e.g. CpG oligonucleotides).
  • Phosphatidylcholine derivatives and phosphorylcholine containing molecules.
  • the invention may also comprise combinations of aspects of one or more of the adjuvants identified above.
  • adjuvant compositions may be used in the invention:
  • the adjuvants described herein can be added to the composition at various stages during their production.
  • the adjuvant may be within or surround an antigen composition, and this mixture can then be/added to an oil-in-water emulsion.
  • the antigen and/adjuvant may be within an oil-in-water emulsion, in which case the agent can either be added to the emulsion components before emulsification, or it can be added to the emulsion after emulsification.
  • the agent may be coaccrvatcd within the emulsion droplets.
  • the location and distribution of the adjuvant within the final composition will depend on its hydrophilic/lipophilic properties e.g . the agent can be located in the aqueous phase, in the oil phase, and/or at the oil-water interface.
  • the adjuvant described herein can be conjugated to a separate agent, such as an antigen (e.g . CRM197) or directly to any amenable composition of the present invention.
  • a separate agent such as an antigen (e.g . CRM197) or directly to any amenable composition of the present invention.
  • conjugation techniques for small molecules is provided in Thompson et al. (2003) Methods in Molecular Medicine 94:255-266 .
  • Preferred conjugation methods involve directly coupling through reductive amination or via a linker, such as adipic acid or squarate.
  • the adjuvants may be non-covalently associated with additional agents, such as by way of hydrophobic or ionic interactions.
  • the invention is also directed to methods of administering the immunogenic compositions of the invention, wherein the immunogenic composition can include in one embodiment one or more adjuvnats and antigens as described herein in combination with a compound of Formula (I) or (II).
  • the immunogenic composition is administered to the subject in an amount effective to stimulate an immune response.
  • the amount that constitutes an effective amount depends, inter alia, on the particular immunogenic composition used, the particular adjuvant compound being administered and the amount thereof, the immune response that is to be enhanced (humoral or cell mediated), the state of the immune system (e.g., suppressed, compromised, stimulated), and the desired therapeutic result. Accordingly it is not practical to set forth generally the amount that constitutes an effective amount of the immunogenic composition. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • compositions of the invention can be administered to various animals subjects including mammals such as human and non-human subjects, including, for example, pocket pets, fowl, and the like according to conventional methods well-known to those skilled in the art.
  • the immunogenic compositions of the present invention can be used in the manufacture of a vaccine.
  • Suitable vaccines include, but are not limited to, any material that raises either or both humoral or cell mediated immune response.
  • Suitable vaccines can include live viral and bacterial antigens and inactivated viral, tumor-derived, protozoal, organism-derived, fungal, and bacterial antigens, toxoids, toxins, polysaccharides, proteins, glycoproteins, peptides, and the like, numerous examples of which are described below.
  • compositions of the invention may be administered in conjunction with one or more antigens for use in therapeutic, prophylactic, or diagnostic methods of the present invention.
  • Preferred antigens include those listed below. Additionally, the compositions of the present invention may be used to treat or prevent infections caused by any of the below-listed pathogens. In addition to combination with the antigens described below, the compositions of the invention may also be combined with an adjuvant as described herein.
  • Antigens for use with the invention include, but are not limited to, one or more of the following antigens set forth below, or antigens derived from one or more of the pathogens set forth below:
  • Bacterial antigens suitable for use in the invention include proteins, polysaccharides, lipopolysaccharides, and outer membrane vesicles which may be isolated, purified or derived from a bacteria.
  • bacterial antigens may include bacterial lysates and inactivated bacteria formulations.
  • Bacteria antigens may be produced by recombinant expression.
  • Bacterial antigens preferably include epitopes which are exposed on the surface of the bacteria during at least one stage of its life cycle. Bacterial antigens are preferably conserved across multiple serotypes.
  • Bacterial antigens include antigens derived from one or more of the bacteria set forth below as well as the specific antigens examples identified below.
  • Meningitides antigens may include proteins (such as those identified in References 1 - 7), saccharides (including a polysaccharide, oligosaccharide or lipopolysaccharide), or outer-membrane vesicles (References 8, 9, 10, 11) purified or derived from N. meningitides serogroup such as A, C, W135, Y, and/or B. Meningitides protein antigens may be selected from adhesions, autotransporters, toxins, Fe acquisition proteins, and membrane associated proteins (preferably integral outer membrane protein).
  • Streptococcus pneumoniae antigens may include a saccharide (including a polysaccharide or an oligosaccharide) and/or protein from Streptococcus pneumoniae. Saccharide antigens may be selected from serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F, and 33F. Protein antigens may be selected from a protein identified in WO 98/18931 , WO 98/18930 , US Patent No. 6,699,703 , US Patent No.
  • Streptococcus pneumoniae proteins may be selected from the Poly Histidine Triad family (PhtX), the Choline Binding Protein family (CbpX), CbpX truncates, LytX family, LytX truncates, CbpX truncate-LytX truncate chimeric proteins, pneumolysin (Ply), PspA, PsaA, Sp128, Sp101, Sp130, Sp125 or Sp133.
  • PhtX Poly Histidine Triad family
  • CbpX Choline Binding Protein family
  • CbpX truncates CbpX truncates
  • LytX family LytX truncates
  • pneumolysin (Ply) PspA, PsaA, Sp128, Sp101, Sp130, Sp125 or Sp133.
  • Streptococcus pyogenes Group A Streptococcus antigens may include a protein identified in WO 02/34771 or WO 2005/032582 (including GAS 40), fusions of fragments of GAS M proteins (including those described in WO 02/094851 , and Dale, Vaccine (1999) 17:193-200 , and Dale, Vaccine 14(10): 944-948 ), fibronectin binding protein (Sfb1), Streptococcal heme-associated protein (Shp), and Streptolysin S (SagA).
  • Moraxella catarrhalis Moraxella antigens include antigens identified in WO 02/18595 and WO 99/58562 , outer membrane protein antigens (HMW-OMP), C-antigen, and/or LPS.
  • HMW-OMP outer membrane protein antigens
  • C-antigen C-antigen
  • LPS LPS
  • Pertussis antigens include pertussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B. pertussis, optionally also combination with pertactin and/or agglutinogens 2 and 3 antigen.
  • PT pertussis holotoxin
  • FHA filamentous haemagglutinin
  • Staphylococcus aureus antigens include S. aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa exotoxin A, such as StaphVAXTM, or antigens derived from surface proteins, invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit phagocytic engulfment (capsule, Protein A), carotenoids, catalase production, Protein A, coagulase, clotting factor, and/or membrane-damaging toxins (optionally detoxified) that lyse eukaryotic cell membranes (hemolysins, leukotoxin, leukocidin).
  • Staph aureus antigens include S. aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa
  • S. epidermidis antigens include slime-associated antigen (SAA).
  • Tetanus antigens include tetanus toxoid (TT), preferably used as a carrier protein in conjunction/conjugated with the compositions of the present invention.
  • TT tetanus toxoid
  • Diphtheria antigens include diphtheria toxin, preferably detoxified, such as CRM 197 . Additionally antigens capable of modulating, inhibiting or associated with ADP ribosylation are contemplated for combination/coadministration/conjugation with the compositions of the present invention.
  • the diphtheria toxoids may be used as carrier proteins.
  • Hib antigens include a Hib saccharide antigen.
  • Pseudomonas aeruginosa Pseudomonas antigens include endotoxin A, Wzz protein, P. aeruginosa LPS, more particularly LPS isolated from PAO1 (O5 serotype), and/or Outer Membrane Proteins, including Outer Membrane Proteins F (OprF) ( Infect Immun. 2001 May; 69(5): 3510-3515 ).
  • Bacterial antigens may be derived from Legionella pneumophila.
  • Streptococcus agalactiae Group B Streptococcus antigens include a protein or saccharide antigen identified in WO 02/34771 , WO 03/093306 , WO 04/041157 , or WO 2005/002619 (including proteins GBS 80, GBS 104, GBS 276 and GBS 322, and including saccharide antigens derived from serotypes Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII).
  • Neiserria gonorrhoeae antigens include Por (or porin) protein, such as PorB ( see Zhu et al., Vaccine (2004) 22:660 - 669 ), a transferring binding protein, such as TbpA and TbpB (See Price et al., Infection and Immunity (2004) 71(1):277 - 283 ), a opacity protein (such as Opa), a reduction-modifiable protein (Rmp), and outer membrane vesicle (OMV) preparations ( see Plante et al., J Infectious Disease (2000) 182:848 - 855 ), also see e.g . WO99/24578 , WO99/36544 , WO99/57280 , WO02/079243 ).
  • PorB see Zhu et al., Vaccine (2004) 22:660 - 669
  • TbpA and TbpB See Price et al., Infection and
  • Chlamydia trachomatis antigens include antigens derived from serotypes A, B, Ba and C (agents of trachoma, a cause of blindness), serotypes L 1 , L 2 & L 3 (associated with Lymphogranuloma venereum), and serotypes, D-K.
  • Chlamydia trachomas antigens may also include an antigen identified in WO 00/37494 , WO 03/049762 , WO 03/068811 , or WO 05/002619 , including PepA (CT045), LcrE (CT089), ArtJ (CT381), DnaK (CT396), CT398, OmpH-like (CT242), L7/L12 (CT316), OmcA (CT444), AtosS (CT467), CT547, Eno (CT587), HrtA (CT823), and MurG (CT761).
  • Treponema pallidum Syphilis antigens include TmpA antigen.
  • Ducreyi antigens include outer membrane protein (DsrA).
  • Antigens include a trisaccharide repeat or other Enterococcus derived antigens provided in US Patent No. 6,756,361 .
  • H pylori antigens include Cag, Vac, Nap, HopX, HopY and/or urease antigen.
  • Antigens include the 160 kDa hemagglutinin of S. saprophyticus antigen.
  • Yersinia enterocolitica Antigens include LPS ( Infect Immun. 2002 August; 70(8): 4414 ).
  • E. coli antigens may be derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), and/or enterohemorrhagic E. coli (EHEC).
  • ETEC enterotoxigenic E. coli
  • EAggEC enteroaggregative E. coli
  • DAEC diffusely adhering E. coli
  • EPEC enteropathogenic E. coli
  • EHEC enterohemorrhagic E. coli
  • Bacillus anthracis Bacillus anthracis (anthrax): B. anthracis antigens are optionally detoxified and may be selected from A-components (lethal factor (LF) and edema factor (EF)), both of which can share a common B-component known as protective antigen (PA).
  • LF lethal factor
  • EF edema factor
  • PA protective antigen
  • Plague antigens include F1 capsular antigen ( Infect Immun. 2003 Jan; 71(1)): 374-383 , LPS ( Infect Immun. 1999 Oct; 67(10): 5395 ), Yersinia pestis V antigen ( Infect Immun. 1997 Nov; 65(11): 4476-4482 ).
  • Tuberculosis antigens include lipoproteins, LPS, BCG antigens, a fusion protein of antigen 85B (Ag85B) and/or ESAT-6 optionally formulated in cationic lipid vesicles ( Infect Immun. 2004 October; 72(10): 6148 ), Mycobacterium tuberculosis (Mtb) isocitrate dehydrogenase associated antigens ( Proc Natl Acad Sci USA. 2004 Aug 24; 101(34):12652 ), and/or MPT51 antigens ( Infect Immun. 2004 July; 72(7 ): 2004 Aug 24; 101(34): 12652 ), and/or MPT51 antigens ( Infect Immun. 2004 July; 72(7): 3829 ).
  • Antigens include outer membrane proteins, including the outer membrane protein A and/or B (OmpB) ( Biochim Biophys Acta. 2004 Nov 1;1702(2):145 ), LPS, and surface protein antigen (SPA) ( J Autoimmun. 1989 Jun;2 Suppl:81 ).
  • OmpB outer membrane protein A and/or B
  • SPA surface protein antigen
  • Bacterial antigens may be derived from Listeria monocytogenes.
  • Chlamydia pneumoniae Antigens include those identified in WO 02/02606 .
  • Antigens include proteinase antigens, LPS, particularly lipopolysaccharides of Vibrio cholerae II, O1 Inaba O-specific polysaccharides, V. cholera 0139, antigens of IEM108 vaccine ( Infect Immun. 2003 Oct;71(10):5498-504 ), and/or Zonula occludens toxin (Zot).
  • Salmonella typhi typhoid fever
  • Antigens include capsular polysaccharides preferably conjugates (Vi, i.e. vax-TyVi).
  • Antigens include lipoproteins (such as OspA, OspB, Osp C and Osp D), other surface proteins such as OspE-related proteins (Erps), decorin-binding proteins (such as DbpA), and antigenically variable VI proteins. , such as antigens associated with P39 and P13 (an integral membrane protein, Infect Immun. 2001 May; 69(5): 3323-3334 ), VlsE Antigenic Variation Protein ( J Clin Microbiol. 1999 Dec; 37(12): 3997 ).
  • Antigens include P. gingivalis outer membrane protein (OMP).
  • Antigens include an OMP, including OMP A, or a polysaccharide optionally conjugated to tetanus toxoid.
  • Further bacterial antigens of the invention may be capsular antigens, polysaccharide antigens or protein antigens of any of the above. Further bacterial antigens may also include an outer membrane vesicle (OMV) preparation. Additionally, antigens include live, attenuated, and/or purified versions of any of the aforementioned bacteria.
  • the antigens of the present invention may be derived from gram-negative or gram-positive bacteria.
  • the antigens of the present invention may be derived from aerobic or anaerobic bacteria.
  • any of the above bacterial-derived saccharides can be conjugated to another agent or antigen, such as a carrier protein (for example CRM 197 ).
  • a carrier protein for example CRM 197
  • Such conjugation may be direct conjugation effected by reductive amination of carbonyl moieties on the saccharide to amino groups on the protein, as provided in US Patent No. 5,360,897 and Can JBiochem Cell Biol. 1984 May;62(5):270-5 .
  • the saccharides can be conjugated through a linker, such as, with succinamide or other linkages provided in Bioconjugate Techniques, 1996 and CRC, Chemistry of Protein Conjugation and Cross-Linking, 1993 .
  • Viral antigens suitable for use in the invention include inactivated (or killed) virus, attenuated virus, split virus formulations, purified subunit formulations, viral proteins which may be isolated, purified or derived from a virus, and Virus Like Particles (VLPs).
  • Viral antigens may be derived from viruses propagated on cell culture or other substrate. Alternatively, viral antigens may be expressed recombinantly.
  • Viral antigens preferably include epitopes which are exposed on the surface of the virus during at least one stage of its life cycle. Viral antigens are preferably conserved across multiple serotypes or isolates. Viral antigens include antigens derived from one or more of the viruses set forth below as well as the specific antigens examples identified below.
  • Orthomyxovirus Viral antigens may be derived from an Orthomyxovirus, such as Influenza A, B and C.
  • Orthomyxovirus antigens may be selected from one or more of the viral proteins, including hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix protein (M1), membrane protein (M2), one or more of the transcriptase components (PB1, PB2 and PA).
  • Preferred antigens include HA and NA.
  • Influenza antigens may be derived from interpandemic (annual) flu strains.
  • influenza antigens may be derived from strains with the potential to cause pandemic a pandemic outbreak (i.e., influenza strains with new haemagglutinin compared to the haemagglutinin in currently circulating strains, or influenza strains which are pathogenic in avian subjects and have the potential to be transmitted horizontally in the human population, or influenza strains which are pathogenic to humans).
  • Viral antigens may be derived from Paramyxoviridae viruses, such as Pneumoviruses (RSV), Paramyxoviruses (PIV) and Morbilliviruses (Measles).
  • RSV Pneumoviruses
  • PMV Paramyxoviruses
  • Measles Morbilliviruses
  • Viral antigens may be derived from a Pneumovirus, such as Respiratory syncytial virus (RSV), Bovine respiratory syncytial virus, Pneumonia virus of mice, and Turkey rhinotracheitis virus.
  • the Pneumovirus is RSV.
  • Pneumovirus antigens may be selected from one or more of the following proteins, including surface proteins Fusion (F), Glycoprotein (G) and Small Hydrophobic protein (SH), matrix proteins M and M2, nucleocapsid proteins N, P and L and nonstructural proteins NS1 and NS2.
  • Preferred Pneumovirus antigens include F, G and M. See e.g., J Gen Virol. 2004 Nov; 85(Pt 11):3229 ).
  • Pneumovirus antigens may also be formulated in or derived from chimeric viruses.
  • chimeric RSV/PIV viruses may comprise components of both RSV and PIV.
  • Viral antigens may be derived from a Paramyxovirus, such as Parainfluenza virus types 1 - 4 (PIV), Mumps, Sendai viruses, Simian virus 5, Bovine parainfluenza virus and Newcastle disease virus.
  • the Paramyxovirus is PIV or Mumps.
  • Paramyxovirus antigens may be selected from one or more of the following proteins: Hemagglutinin -Neuraminidase (HN), Fusion proteins F1 and F2, Nucleoprotein (NP), Phosphoprotein (P), Large protein (L), and Matrix protein (M).
  • HN Hemagglutinin -Neuraminidase
  • NP Nucleoprotein
  • Phosphoprotein P
  • Large protein L
  • Matrix protein M
  • Preferred Paramyxovirus proteins include HN, F1 and F2.
  • Paramyxovirus antigens may also be formulated in or derived from chimeric viruses.
  • chimeric RSV/PIV viruses may comprise components of both RSV and PIV.
  • Commercially available mumps vaccines include live attenuated mumps virus, in either a monovalent form or in combination with measles and rubella vaccines (MMR).
  • Morbillivirus Viral antigens may be derived from a Morbillivirus, such as Measles. Morbillivirus antigens may be selected from one or more of the following proteins: hemagglutinin (H), Glycoprotein (G), Fusion factor (F), Large protein (L), Nucleoprotein (NP), Polymerase phosphoprotein (P), and Matrix (M).
  • H hemagglutinin
  • G Glycoprotein
  • F Fusion factor
  • L Large protein
  • NP Nucleoprotein
  • P Polymerase phosphoprotein
  • M Matrix
  • Commercially available measles vaccines include live attenuated measles virus, typically in combination with mumps and rubella (MMR).
  • Viral antigens may be derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and Aphthoviruses. Antigens derived from Enteroviruses, such as Poliovirus are preferred.
  • Viral antigens may be derived from an Enterovirus, such as Poliovirus types 1, 2 or 3, Coxsackie A virus types 1 to 22 and 24, Coxsackie B virus types 1 to 6, Echovirus (ECHO) virus) types 1 to 9, 11 to 27 and 29 to 34 and Enterovirus 68 to 71.
  • the Enterovirus is poliovirus.
  • Enterovirus antigens are preferably selected from one or more of the following Capsid proteins VP1, VP2, VP3 and VP4.
  • Commercially available polio vaccines include Inactivated Polio Vaccine (IPV) and Oral poliovirus vaccine (OPV).
  • Heparnavirus Viral antigens may be derived from an Heparnavirus, such as Hepatitis A virus (HAV).
  • HAV Hepatitis A virus
  • Commercially available HAV vaccines include inactivated HAV vaccine.
  • Viral antigens may be derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. Antigens derived from Rubivirus, such as Rubella virus, are preferred. Togavirus antigens may be selected from E1, E2, E3, C, NSP-1, NSPO-2, NSP-3 or NSP-4. Togavirus antigens are preferably selected from E1, E2 or E3.
  • Commercially available Rubella vaccines include a live cold-adapted virus, typically in combination with mumps and measles vaccines (MMR).
  • Flavivirus Viral antigens may be derived from a Flavivirus, such as Tick-borne encephalitis (TBE), Dengue (types 1, 2, 3 or 4), Yellow Fever, Japanese encephalitis, West Nile encephalitis, St. Louis encephalitis, Russian spring-summer encephalitis, Powassan encephalitis. Flavivirus antigens may be selected from PrM, M, C, E, NS-1, NS-2a, NS2b, NS3, NS4a, NS4b, and NS5. Flavivirus antigens are preferably selected from PrM, M and E. Commercially available TBE vaccine include inactivated virus vaccines.
  • TBE vaccine include inactivated virus vaccines.
  • Pestivirus Viral antigens may be derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).
  • BVDV Bovine viral diarrhea
  • CSFV Classical swine fever
  • BDV Border disease
  • Hepadnavirus Viral antigens may be derived from a Hepadnavirus, such as Hepatitis B virus. Hepadnavirus antigens may be selected from surface antigens (L, M and S), core antigens (HBc, HBe). Commercially available HBV vaccines include subunit vaccines comprising the surface antigen S protein.
  • Hepatitis C virus Viral antigens may be derived from a Hepatitis C virus (HCV). ( see, e.g . Hsu et al. (1999) Clin Liver Dis 3:901-915 ). HCV antigens may be selected from one or more of E1, E2, E1/E2, NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions ( Houghton et al., Hepatology (1991) 14:381 ).
  • HCV antigens may be selected from one or more of E1, E2, E1/E2, NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions ( Houghton et al., Hepatology (1991) 14:381 ).
  • Hepatitis C virus antigens that may be used can include one or more of the following: HCV E1 and or E2 proteins, E1/E2 heterodimer complexes, core proteins and non-structural proteins, or fragments of these antigens, wherein the non-structural proteins can optionally be modified to remove enzymatic activity but retain immunogenicity ( see, e.g . WO03/002065 ; WO01/37869 and WO04/005473 ).
  • Viral antigens may be derived from a Rhabdovirus, such as a Lyssavirus (Rabies virus) and Vesiculovirus (VSV).
  • Rhabdovirus antigens may be selected from glycoprotein (G), nucleoprotein (N), large protein (L), nonstructural proteins (NS).
  • G glycoprotein
  • N nucleoprotein
  • L large protein
  • NS nonstructural proteins
  • Rabies virus vaccine comprise killed virus grown on human diploid cells or fetal rhesus lung cells.
  • Viral antigens may be derived from Calciviridae, such as Norwalk virus, and Norwalk-like Viruses, such as Hawaii Virus and Snow Mountain Virus.
  • Coronavirus Viral antigens may be derived from a Coronavirus, SARS, Human respiratory coronavirus, Avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV). Coronavirus antigens may be selected from spike (S), envelope (E), matrix (M), nucleocapsid (N), and Hemagglutininesterase glycoprotein (HE). Preferably, the Coronavirus antigen is derived from a SARS virus. SARS viral antigens are described in WO 04/92360 ;
  • Retrovirus Viral antigens may be derived from a Retrovirus, such as an Oncovirus, a Lentivirus or a Spumavirus.
  • Oncovirus antigens may be derived from HTLV-1, HTLV-2 or HTLV-5.
  • Lentivirus antigens may be derived from HIV-1 or HIV-2.
  • Retrovirus antigens may be selected from gag, pol, env, tax, tat, rex, rev, nef, vif, vpu, and vpr.
  • HIV antigens may be selected from gag (p24gag and p55gag), env (gp160 and gp41), pol, tat, nef, rev vpu, miniproteins, (preferably p55 gag and gp140v delete). HIV antigens may be derived from one or more of the following strains: HIV IIIb , HIV SF2 , HIV LAV , HIV LAI , HIV MN , HIV-1 CM235 , HIV-1 US4 .
  • Viral antigens may be derived from a Reovirus, such as an Orthoreovirus, a Rotavirus, an Orbivirus, or a Coltivirus.
  • Reovirus antigens may be selected from structural proteins ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 1, ⁇ 2, ⁇ 1, ⁇ 2, or ⁇ 3, or nonstructural proteins ⁇ NS, ⁇ NS, or ⁇ 1s.
  • Preferred Reovirus antigens may be derived from a Rotavirus.
  • Rotavirus antigens may be selected from VP1, VP2, VP3, VP4 (or the cleaved product VP5 and VP8), NSP 1, VP6, NSP3, NSP2, VP7, NSP4, or NSP5.
  • Preferred Rotavirus antigens include VP4 (or the cleaved product VP5 and VP8), and VP7.
  • Viral antigens may be derived from a Parvovirus, such as Parvovirus B19. Parvovirus antigens may be selected from VP-1, VP-2, VP-3, NS-1 and NS-2. Preferably, the Parvovirus antigen is capsid protein VP-2.
  • Delta hepatitis virus Viral antigens may be derived HDV, particularly ⁇ -antigen from HDV (see, e.g., U.S. Patent No. 5,378,814 ).
  • HEV Hepatitis E virus
  • Hepatitis G virus Viral antigens may be derived from HGV.
  • Human Herpesvirus Viral antigens may be derived from a Human Herpesvirus, such as Herpes Simplex Viruses (HSV), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human Herpesvirus 8 (HHV8).
  • Human Herpesvirus antigens may be selected from immediate early proteins ( ⁇ ), early proteins ( ⁇ ), and late proteins ( ⁇ ).
  • HSV antigens may be derived from HSV-1 or HSV-2 strains.
  • HSV antigens may be selected from glycoproteins gB, gC, gD and gH, fusion protein (gB), or immune escape proteins (gC, gE, or gI).
  • VZV antigens may be selected from core, nucleocapsid, tegument, or envelope proteins.
  • a live attenuated VZV vaccine is commercially available.
  • EBV antigens may be selected from early antigen (EA) proteins, viral capsid antigen (VCA), and glycoproteins of the membrane antigen (MA).
  • CMV antigens may be selected from capsid proteins, envelope glycoproteins (such as gB and gH), and tegument proteins
  • Papovaviruses Antigens may be derived from Papovaviruses, such as Papillomaviruses and Polyomaviruses.
  • Papillomaviruses include HPV serotypes 1, 2, 4, 5, 6, 8, 11, 13, 16, 18, 31, 33, 35, 39, 41, 42, 47, 51, 57, 58, 63 and 65.
  • HPV antigens are derived from serotypes 6, 11, 16 or 18.
  • HPV antigens may be selected from capsid proteins (L1) and (L2), or E1 - E7, or fusions thereof.
  • HPV antigens are preferably formulated into virus-like particles (VLPs).
  • Polyomyavirus viruses include BK virus and JK virus.
  • Polyomavirus antigens may be selected from VP1, VP2 or VP3.
  • Vaccines 4th Edition
  • Medical Microbiology 4th Edition (Murray et al. ed. 2002 ); Virology, 3rd Edition (W.K. Joklik ed. 1988 ); Fundamental Virology, 2nd Edition (B.N. Fields and D.M. Knipe, eds. 1991 ), which are contemplated in conjunction with the compositions of the present invention.
  • Fungal antigens for use in the invention may be derived from one or more of the fungi set forth below.
  • Fungal antigens may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var. discoides, var. ochraceum, Trichophyton violaceum, and/or Trichophyton faviforme.
  • Dermatophytres including: Epidermophyton floccusum, Microsporum audouini,
  • Fungal pathogens may be derived from Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida parapsilosis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladosporium carrionii, Coccidioides immitis, Blastomyces dermatidis, Cryptococcus neoformans, Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Paracoccidioides brasi
  • a solubilized fraction extracted and separated from an insoluble fraction obtainable from fungal cells of which cell wall has been substantially removed or at least partially removed characterized in that the process comprises the steps of: obtaining living fungal cells; obtaining fungal cells of which cell wall has been substantially removed or at least partially removed; bursting the fungal cells of which cell wall has been substantially removed or at least partially removed; obtaining an insoluble fraction; and extracting and separating a solubilized fraction from the insoluble fraction.
  • compositions of the invention may include one or more antigens derived from a sexually transmitted disease (STD).
  • STD sexually transmitted disease
  • Such antigens may provide for prophylactis or therapy for STD's such as chlamydia, genital herpes, hepatits (such as HCV), genital warts, gonorrhoea, syphilis and/or chancroid (See, WO00/15255 ).
  • Antigens may be derived from one or more viral or bacterial STD's.
  • Viral STD antigens for use in the invention may be derived from, for example, HIV, herpes simplex virus (HSV-1 and HSV-2), human papillomavirus (HPV), and hepatitis (HCV).
  • Bacterial STD antigens for use in the invention may be derived from, for example, Neiserria gonorrhoeae, Chlamydia trachomatis, Treponema pallidum, Haemophilus ducreyi, E. coli, and Streptococcus agalactiae. Examples of specific antigens derived from these pathogens are described above.
  • compositions of the invention may include one or more antigens derived from a pathogen which causes respiratory disease.
  • respiratory antigens may be derived from a respiratory virus such as Orthomyxoviruses (influenza), Pneumovirus (RSV), Paramyxovirus (PIV), Morbillivirus (measles), Togavirus (Rubella), VZV, and Coronavirus (SARS).
  • Respiratory antigens may be derived from a bacteria which causes respiratory disease, such as Streptococcus pneumoniae, Pseudomonas aeruginosa, Bordetella pertussis, Mycobacterium tuberculosis, Mycoplasma pneumoniae, Chlamydia pneumoniae, Bacillus anthracis, and Moraxella catarrhalis. Examples of specific antigens derived from these pathogens are described above.
  • compositions of the invention may include one or more antigens suitable for use in pediatric subjects.
  • Pediatric subjects are typically less than about 3 years old, or less than about 2 years old, or less than about 1 years old.
  • Pediatric antigens may be administered multiple times over the course of 6 months, 1, 2 or 3 years.
  • Pediatric antigens may be derived from a virus which may target pediatric populations and/or a virus from which pediatric populations are susceptible to infection.
  • Pediatric viral antigens include antigens derived from one or more of Orthomyxovirus (influenza), Pneumovirus (RSV), Paramyxovirus (PIV and Mumps), Morbillivirus (measles), Togavirus (Rubella), Enterovirus (polio), HBV, Coronavirus (SARS), and Varicella-zoster virus (VZV), Epstein Barr virus (EBV).
  • Orthomyxovirus influenza
  • RSV Pneumovirus
  • PIV and Mumps Paramyxovirus
  • Morbillivirus measles
  • Togavirus Rubella
  • Enterovirus polio
  • HBV HBV
  • Coronavirus Coronavirus
  • VZV Varicella-zoster virus
  • EBV Epstein Barr virus
  • Pediatric bacterial antigens include antigens derived from one or more of Streptococcus pneumoniae, Neisseria meningitides, Streptococcus pyogenes (Group A Streptococcus), Moraxella catarrhalis, Bordetella pertussis, Staphylococcus aureus, Clostridium tetani (Tetanus), Cornynebacterium diphtheriae (Diphtheria), Haemophilus influenzae B (Hib), Pseudomonas aeruginosa, Streptococcus agalactiae (Group B Streptococcus), and E. coli. Examples of specific antigens derived from these pathogens are described above.
  • compositions of the invention may include one or more antigens suitable for use in elderly or immunocompromised individuals. Such individuals may need to be vaccinated more frequently, with higher doses or with adjuvanted formulations to improve their immune response to the targeted antigens.
  • Antigens which may be targeted for use in Elderly or Immunocompromised individuals include antigens derived from one or more of the following pathogens: Neisseria meningitides, Streptococcus pneumoniae, Streptococcus pyogenes (Group A Streptococcus), Moraxella catarrhalis, Bordetella pertussis, Staphylococcus aureus, Staphylococcus epidermis, Clostridium tetani (Tetanus), Cornynebacterium diphtheriae (Diphtheria), Haemophilus influenzae B (Hib), Pseudomonas aeruginosa, Legionella pneumophila, Streptococcus
  • compositions of the invention may include one or more antigens suitable for use in adolescent subjects.
  • Adolescents may be in need of a boost of a previously administered pediatric antigen.
  • Pediatric antigens which may be suitable for use in adolescents are described above.
  • adolescents may be targeted to receive antigens derived from an STD pathogen in order to ensure protective or therapeutic immunity before the beginning of sexual activity.
  • STD antigens which may be suitable for use in adolescents are described above.
  • Tumor antigens can be, for example, peptide-containing tumor antigens, such as a polypeptide tumor antigen or glycoprotein tumor antigens.
  • a tumor antigen can also be, for example, a saccharide-containing tumor antigen, such as a glycolipid tumor antigen or a ganglioside tumor antigen.
  • the tumor antigen can further be, for example, a polynucleotide-containing tumor antigen that expresses a polypeptide-containing tumor antigen, for instance, an RNA vector construct or a DNA vector construct, such as plasmid DNA.
  • Tumor antigens appropriate for the practice of the present invention encompass a wide variety of molecules, such as (a) polypeptide-containing tumor antigens, including polypeptides (which can range, for example, from 8-20 amino acids in length, although lengths outside this range are also common), lipopolypeptides and glycoproteins, (b) saccharide-containing tumor antigens, including poly-saccharides, mucins, gangliosides, glycolipids and glycoproteins, and (c) polynucleotides that express antigenic polypeptides.
  • the tumor antigens can be, for example, (a) full length molecules associated with cancer cells, (b) homologs and modified forms of the same, including molecules with deleted, added and/or substituted portions, and (c) fragments of the same.
  • Tumor antigens can be provided in recombinant form.
  • Tumor antigens include, for example, class I-restricted antigens recognized by CD8+ lymphocytes or class II-restricted antigens recognized by CD4+ lymphocytes.
  • tumor antigens are known in the art, including: (a) cancer-testis antigens such as NY-ESO-1, SSX2, SCP1 as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-1, GAGE-2, MAGE-1, MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors), (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUM1 (associated with, e.g., melanoma), caspase-8 (associated cancer
  • Additional tumor antigens which are known in the art include p15, Hom/Mel-40, H-Ras, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, p185erbB2, p180erbB-3, c-met, mn-23H1, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, p16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO-029
  • Polynucleotide-containing antigens in accordance with the present invention typically comprise polynucleotides that encode polypeptide cancer antigens such as those listed above.
  • Preferred polynucleotide-containing antigens include DNA or RNA vector constructs, such as plasmid vectors (e.g., pCMV), which are capable of expressing polypeptide cancer antigens in vivo.
  • Tumor antigens may be derived, for example, from mutated or altered cellular components. After alteration, the cellular components no longer perform their regulatory functions, and hence the cell may experience uncontrolled growth.
  • altered cellular components include ras, p53, Rb, altered protein encoded by the Wilms' tumor gene, ubiquitin, mucin, protein encoded by the DCC, APC, and MCC genes, as well as receptors or receptor-like structures such as neu, thyroid hormone receptor, platelet derived growth factor (PDGF) receptor, insulin receptor, epidermal growth factor (EGF) receptor, and the colony stimulating factor (CSF) receptor.
  • PDGF platelet derived growth factor
  • EGF epidermal growth factor
  • CSF colony stimulating factor
  • bacterial and viral antigens may be used in conjunction with the compositions of the present invention for the treatment of cancer.
  • carrier proteins such as CRM 197 , tetanus toxoid, or Salmonella typhimurium antigen can be used in conjunction/conjugation with compounds of the present invention for treatment of cancer.
  • the cancer antigen combination therapies will show increased efficacy and bioavailability as compared with existing therapies.
  • methods of producing microparticles having adsorbed antigens comprise: (a) providing an emulsion by dispersing a mixture comprising (i) water, (ii) a detergent, (iii) an organic solvent, and (iv) a biodegradable polymer selected from the group consisting of a poly( ⁇ -hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate.
  • the polymer is typically present in the mixture at a concentration of about 1% to about 30% relative to the organic solvent, while the detergent is typically present in the mixture at a weight-to-weight detergent-to-polymer ratio of from about 0.00001:1 to about 0.1:1 (more typically about 0.0001:1 to about 0.1:1, about 0.001:1 to about 0.1:1, or about 0.005:1 to about 0.1:1); (b) removing the organic solvent from the emulsion; and (c) adsorbing an antigen on the surface of the microparticles.
  • the biodegradable polymer is present at a concentration of about 3% to about 10% relative to the organic solvent.
  • Microparticles for use herein will be formed from materials that are sterilizable, non-toxic and biodegradable. Such materials include, without limitation, poly( ⁇ -hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate.
  • microparticles for use with the present invention are derived from a poly( ⁇ -hydroxy acid), in particular, from a poly(lactide) ("PLA”) or a copolymer of D,L-lactide and glycolide or glycolic acid, such as a poly(D,L-lactide-co-glycolide) (“PLG” or "PLGA”), or a copolymer of D,L-lactide and caprolactone.
  • PLA poly(lactide)
  • PLA poly(lactide)
  • PLA poly(D,L-lactide-co-glycolide)
  • caprolactone a copolymer of D,L-lactide and caprolactone
  • microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of which will be largely a matter of choice, depending in part on the coadministered macromolecule. These parameters are discussed more fully below.
  • antigens may also include an outer membrane vesicle (OMV) preparation.
  • OMV outer membrane vesicle
  • compositions that include the compounds described herein may include additives such as excipients.
  • suitable pharmaceutically acceptable excipients include processing agents and drug delivery modifiers and enhancers, such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, hydroxypropyl- ⁇ -cyclodextrin, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or more of these.
  • Other suitable pharmaceutically acceptable excipients are described in " Remington's Pharmaceutical Sciences," Mack Pub. Co., New Jersey (1991 ), which is hereby incorporated herein by reference in its entirety and for all purposes as if fully set forth herein.
  • compositions that include the compounds of the invention may be in any form suitable for the intended method of administration, including, for example, as a solution, a suspension, or an emulsion.
  • Liquid carriers are typically used in preparing solutions, suspensions, and emulsions.
  • Liquid carriers contemplated for use in the practice of the present invention include, for example, water, saline, pharmaceutically acceptable organic solvent(s), pharmaceutically acceptable oils or fats, and the like, as well as mixtures of two or more of these.
  • the liquid carrier may include other suitable pharmaceutically acceptable additives such as solubilizers, emulsifiers, nutrients, buffers, preservatives, suspending agents, thickening agents, viscosity regulators, stabilizers, and the like.
  • Suitable organic solvents include, for example, monohydric alcohols, such as ethanol, and polyhydric alcohols, such as glycols.
  • Suitable oils include, but are not limited to, soybean oil, coconut oil, olive oil, safflower oil, cottonseed oil, and the like.
  • the carrier may be an oily ester such as ethyl oleate, isopropyl myristate, and the like.
  • Compositions of the present invention may also be in the form of microparticles, microcapsules, and the like, as well as combinations of any two or more of these.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form may include, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like.
  • Preferred lipids include phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods of forming liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.W., p. 33 et seq (1976 ).
  • Controlled release delivery systems may also be used, such as a diffusion controlled matrix system or an erodible system, as described for example in: Lee, "Diffusion-Controlled Matrix Systems", pp. 155-198 and Ron and Langer, “Erodible Systems", pp. 199-224 , in “ Treatise on Controlled Drug Delivery", A. Kydonieus Ed., Marcel Dekker, Inc., New York 1992 .
  • the matrix may be, for example, a biodegradable material that can degrade spontaneously in situ and in vivo for, example, by hydrolysis or enzymatic cleavage, e.g., by proteases.
  • the delivery system may be, for example, a naturally occurring or synthetic polymer or copolymer, for example in the form of a hydrogel.
  • exemplary polymers with cleavable linkages include polyesters, polyorthoesters, polyanhydrides, polysaccharides, poly(phosphoesters), polyamides, polyurethanes, poly(imidocarbonates) and poly(phosphazenes).
  • the compounds of the invention may be administered enterally, orally, parenterally, sublingually, intradermally, by inhalation spray, rectally, or topically in dosage unit formulations that include conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired.
  • suitable modes of administration include oral, subcutaneous, transdermal, transmucosal, iontophoretic, intravenous, intramuscular, intraperitoneal, intranasal, subdermal, rectal, and the like.
  • Topical administration may also include the use of transdermal administration such as transdermal patches or ionophoresis devices.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques.
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-propanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • a suitable nonirritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch.
  • Such dosage forms may also include, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • the dosage forms may also include buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water.
  • Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, cyclodextrins, and sweetening, flavoring, and perfuming agents.
  • Effective amounts of the compounds of the invention generally include any amount sufficient to detectably treat the disorders described herein.
  • Successful treatment of a subject in accordance with the invention may result in a reduction or alleviation of symptoms in a subject afflicted with a medical or biological disorder.
  • treatment may halt the further progression of the disorder, or may prevent or retard development of the disorder.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and severity of the particular disease undergoing therapy. The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
  • SMIP refers to a small molecule immunopotentiating compound, including small molecule compounds, generally below about MW 800 g/mol, capable of stimulating or modulating a pro-inflammatory response in a patient.
  • the SMIP compounds are able to stimulate human peripheral blood mononuclear cells to produce cytokines. More particularly, preferred SMIPs include imidazoquinolines and those compounds encompassed by Formulas (I)a nd (II) described herein, or contained within any reference cited herein.
  • SMIS refers to a small molecule immunosuppressant compound, including small molecule compounds, generally below about MW 800 g/mol, capable of suppressing or modulating an immune response in a patient.
  • the SMIS compounds are able to inhibit human peripheral blood mononuclear cell's ability to produce cytokines, chemokines, and/or growth factors.
  • the SMIS compounds are able to induce TGF-beta production, thereby suppressing an immune response.
  • imidazoquinoxalines (as pertaining to imidazoquinoxalincs of the present invention), indicates compounds imidazo[1,2-a]quinoxalines having the general structure of Formula (I) or (II) as described herein.
  • Some preferred imidazoquinoxalines include compound of Formula:
  • refractory cancer cells refers to cancer cell lines that are resistant to preexisting therapeutics or treatment regimens, including prescribed dosing schedules.
  • the methods of the invention are useful in treating "allergic diseases," which may be accomplished in the same manner as the other immunotherapeutic methods described herein.
  • allergen refers to a substance (antigen) that can induce an allergic or asthmatic response in a susceptible subject.
  • the list of allergens is enormous and can include pollens, insect venoms, animal dander, dust, fungal spores, and drugs (e.g., penicillin).
  • Asthma refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways, and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively, associated with atopic or allergic symptoms.
  • leukotriene inhibitor includes any agent or compound that inhibits, restrains, retards, or otherwise interacts with the action or activity of leukotrienes, such as, but not limited to, 5-lipoxygenase (“5-LO”) inhibitors, 5-lipoxygenase activating protein (“FLAP”) antagonists, and leukotriene D4 (“LTD4 ”) antagonists.
  • 5-LO 5-lipoxygenase
  • FLAP 5-lipoxygenase activating protein
  • LTD4 leukotriene D4
  • Modeulating refers to inducing or suppressing.
  • Immuno-stimulation “immune response” or “immune potentiation” refer to enhancement or activation of the immune system, including humoral or cellular activation, for example, activation of a cell, such as a killer (T or NK) or dendritic cell of the immune system, for example, causing the increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • a cell such as a killer (T or NK) or dendritic cell of the immune system, for example, causing the increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • Modeulating an immune response refers to either immune potentiation or immune suppression as defined herein.
  • an “immunogenic composition” refers to a composition capable of stimulating an immune response.
  • “immunogenic compositions” are compositions capable of stimulating an immune response in a subject.
  • the immunogenic composition is capable of modulating the production of cytokines in a subject, thereby effecting immune potentiation in that subject.
  • Immunosuppression refers to deactivation of the immune system, for example, preventing or lessening cytokine production from a dendritic cell leading to an overall attenuation of host defense (immune response).
  • an “immune-stimulatory effective amount” is an amount effective for activating the immune system, for example, causing an increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • Enhancing the immune response to an antigen by a compound refers to enhancement of the immune response in comparison to that in the absence of the compound.
  • An enhanced immune-response eliciting composition is a composition generally comprising an antigen and a small molecule immune potentiator compound that elicits an immune response greater than a composition comprising an antigen and not containing one or more small molecule immune potentiator compounds.
  • the compound acts as an adjuvant, for example, for use in vaccine compositions and methods.
  • a “disease associated with cellular proliferation” includes, but is not limited to ncuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogcncsis, and endotoxic shock.
  • PDR proliferative diabetic retinopathy
  • an effective amount of a compound to treat an infectious disorder may be an amount necessary to cause an antigen specific immune response upon exposure to an infectious agent.
  • the effective amount may vary, depending, for example, upon the condition treated, weight of the subject and severity of the disease. One of skill in the art can readily determine the effective amount empirically without undue experimentation.
  • an effective amount for treatment refers to an amount sufficient to palliate, ameliorate, stabilize, reverse, slow or delay progression of a condition such as a disease state.
  • Metronomic administration or “administered metronomically” refers to increasingly frequent dosing regimens, at lower drug concentrations, as compared with known dosing regimens for an existing therapeutic. Metronomic administration varies from the typical dosing of cytotoxic drugs, which involves episodic (less frequent) administration at maximum tolerated doses (MTDs).
  • MTDs maximum tolerated doses
  • a “subject” or “patient” is meant to describe a human or vertebrate animal including a dog, cat, pocket pet, marmoset, horse, cow, pig, sheep, goat, elephant, giraffe, chicken, lion, monkey, owl, rat, squirrel, slender loris, and mouse.
  • a “pocket pet” refers to a group of vertebrate animals capable of fitting into a commodious coat pocket such as, for example, hamsters, chinchillas, ferrets, rats, guinea pigs, gerbils, rabbits and sugar gliders. Further description is provided by Mackay, B., Pocket Pets, Animal Issues, 32(1) 2001 .
  • ester refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkancdioic acids, in which each alkyl or alkenyl moicty advantageously has not more than 6 carbon atoms.
  • Representative examples of particular esters include, but arc not limited to, formates, acetates, propionates, butyrates, acrylatcs and ethylsuccinates.
  • the compounds of the present invention can be used in the form of salts as in "pharmaceutically acceptable salts" derived from inorganic or organic acids.
  • These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-napthalenesulfonate, oxalate, pamoate, pe
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such
  • prodrugs refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series , and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987 . Prodrugs as described in U.S. Patent No. 6,284,772 for example may be used.
  • the symbol is meant to indicate the point of attachment of an appendage.
  • halo refers to F, Cl, Br, or I atoms, especially F, Cl, and Br.
  • alkyl refers to groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • C 1-6 alkyl has the same meaning as alkyl, except that it is limited to alkyl groups of six carbons or less.
  • the phrase C 1-6 alkyl. also includes branched chain isomers of straight chain alkyl.
  • C 1 - 6 alkyl further includes cyclic alkyl or C 3-6 cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and such rings substituted with straight and branched chain alkyl groups as defined above.
  • alkyl also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • aryl refers to groups that do not contain heteroatoms.
  • C 6-10 aryl has the same meaning as aryl, except that it is limited to aryl groups of six to ten carbons atoms.
  • aryl includes, but is not limited to, groups such as phenyl, biphenyl, and naphthyl by way of example.
  • Aryl groups also include those in which one of the aromatic carbons is bonded to an alkyl, alkenyl, or alkynyl group as defined herein.
  • aryl includes, but is not limited to tolyl, and hydroxyphenyl among others.
  • alkenyl refers to straight chain, branched chain, and cyclic groups such as those described with respect to alkyl groups as defined above, except that at least one double bond exists between two carbon atoms.
  • C 2-6 alkenyl has the same meaning as alkenyl, except that it is limited to alkenyl groups of two to six carbons.
  • alkoxy refers to groups having the formula -O-alkyl, wherein the point of attachment is the oxy group and the alkyl group is as defined above.
  • C 1-6 alkoxy has the same meaning as alkoxy, except that it is limited to alkoxy groups having from one to six carbon atoms.
  • aryloxy refers to groups having the formula -O-aryl, wherein the point of attachment is the oxy group and the aryl group is as defined above.
  • C 6-10 aryloxy has the same meaning as aryloxy, except that it is limited to aryloxy groups of six to ten carbon atoms.
  • C 1-6 alkoxy-C 1-6 alkyl refers to ether groups with as many as 12 carbon atoms.
  • One example of a C 1-6 alkoxy-C 1-6 alkyl group is -CH 2 -O-CH 2 CH 3 .
  • C 6-10 aryloxy-C 1-6 alkyl refers to aryl ether groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the C 1-6 alkyl group.
  • One example of a C 6-10 aryloxy-C 1-6 alkyl group is propoxybenzene.
  • C 6-10 aryl-C 1-6 alkyl refers to arylalkyl groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the C 1-6 alkyl group.
  • One example of a C 6-10 aryl-C 1-6 alkyl group is toluene.
  • amino refers to NH 2 .
  • alkynyl refers to straight and branched chain groups such as those described with respect to alkyl groups as defined above, except that at least one triple bond exists between two carbon atoms.
  • C 2-6 alkynyl has the same meaning as alkynyl, except that it is limited to alkynyl groups of two to six carbons. Examples include, but are not limited to, -C ⁇ C(H), -C ⁇ C(CH 3 ), -C ⁇ C(CH 2 CH 3 ), -C(H 2 )C ⁇ C(H), -C(H) 2 C ⁇ C(CH 3 ), -C(H) 2 C ⁇ C(CH 2 CH 3 ), and the like.
  • trihalomethyl refers to a methyl group in which the three H atoms of the methyl group are substituted with three halogens which may be same or different.
  • a -CF 3 group is a group in which all three H atoms of the methyl group are substituted with F atoms.
  • -CH 2 C(CH 3 ) 2 (OH) refers to 2-methylpropan-2-ol or tertbutanol.
  • heterocyclyl refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g. 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl etc.), tetrazolyl, (e.g.
  • saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms such as, but not limited to furanyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl (e.g.
  • unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g.
  • 1,3-benzodioxoyl, etc. unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing an oxygen atom and 1 to 2 oxygen atoms such as benzoxathiinyl.
  • unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl
  • saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane
  • Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones).
  • heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1-dioxide.
  • Preferred heterocyclyl groups contain 5 or 6 ring members.
  • More preferred heterocyclyl groups include morpholine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran.
  • Heterocyclyl also refers to those groups as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2-methylbenzimidazolyl, 5-methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1-methyl piperazinyl, and 2-chloropyridyl among others. Heterocyclyl groups are those limited to having 2 to 15 carbon atoms and as many as 6 additional heteroatoms as described above. More preferred heterocyclyl groups have from 3 to 5 carbon atoms and as many as 2 heteroatoms. Most preferred heterocyclyl groups include piperidinyl, pyrrolidinyl, azetidinyl, and aziridinyl groups.
  • substitution groups include, for example, hydroxy, nitro, amino, imino, cyano, halo, thio, thioamido, amidino, imidino, oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamido, carboxyl, formyl, alkyl, heterocyclyl, aryl, haloalkyl, alkoxy, alkoxyalkyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkylthio, aminoalkyl, alkylamino, cyanoalkyl, and the like.
  • substituted C 1-6 alkyl is tertbutanol.
  • Other preferred substituted C 1-6 alkyl groups include alkoxyalkyl groups (i.e., groups of formula -alkyl-O-alkyl), and - CH 2 C(CH 3 ) 2 NH-SO 2 CH 3 .
  • the substitution group can itself be substituted one time.
  • an alkoxy substituent of an alkyl group may be substituted with a halogen, and oxo group, an aryl group, or the like.
  • the group substituted onto the substitution group can be carboxyl, halo, nitro, oxo, amino, cyano, hydroxy, C 1-6 alkyl, C 1-6 alkoxy, C 6-10 aryl, aminocarbonyl, -SR, thioamido, -SO 3 H, -SO 2 R or cycloalkyl, where R is typically hydrogen, hydroxy or C 1-6 alkyl.
  • substituted substituent when the substituted substituent includes a straight chain group, the substitution can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and the like) or at the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like).
  • Substituted substituents can be straight chain, branched or cyclic arrangements of covalently bonded carbon atoms or heteroatoms.
  • protected or a “protecting group” with respect to hydroxy groups, aminc groups, and sulfhydryl groups refers to forms of these functionalities which are protected from undesirable reaction with a protecting group known to those skilled in the art such as those set forth in Protective Groups in Organic Synthesis, Greene, T.W., John Wiley & Sons, New York, NY, (1st Edition, 1981 ) which can be added or removed using the procedures set forth therein.
  • Examples of protected hydroxy groups include, but are not limited to, silyl ethers such as those obtained by reaction of a hydroxy group with a reagent such as, but not limited to, t-butyldimethyl-chlorosilane, trimethylchlorosilane, triisopropylchlorosilane, triethylchlorosilane; substituted methyl and ethyl ethers such as, but not limited to methoxymethyl ether, methythiomethyl ether, benzyloxymethyl ether, t-butoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyranyl ethers, 1-ethoxyethyl ether, allyl ether, benzyl ether; esters such as, but not limited to, benzoylformate, formate, acetate, trichloroacetate, and trifluoracetate.
  • a reagent such as, but not limited to,
  • protected amine groups include, but are not limited to, benzyl or dibenzyl, amides such as, formamide, acetamide, trifluoroacetamide, and benzamide; imides, such as phthalimide, and dithiosuccinimide; and others.
  • a protecting group for amines is a benzyl group.
  • protected sulfhydryl groups include, but are not limited to, thioethers such as S-benzyl thioether, and S-4-picolyl thioether; substituted S-methyl derivatives such as hemithio, dithio and aminothio acetals; and others.
  • Imidazoquinoxaline compounds of Formula (I) or (II) and the analogs thereof may exhibit the phenomenon of tautomerism, and the formula drawings within this specification can represent only one of the possible tautomeric forms. It is to be understood that the invention encompasses any tautomeric form which possesses immunomodulatory activity and is not to be limited merely to any one tautomeric form utilized within the formula drawings.
  • Imidazoquinoxalines of Formula (I) or (II) also may exist in solvated as well as unsolvated forms such as, for example, hydrated forms.
  • the invention encompasses both solvated and unsolvated forms which possess immunomodulatory activity.
  • the invention also includes isotopically-labeled imidazoquinoxaline compounds and analogs thereof, that are structurally identical to those disclosed above, except that one or more atom is/are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 O 17 O, 31 P, 32 P , 35 S 18 F and 36 Cl, respectively.
  • isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out known or referenced procedures and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • Example compounds and their analogs are easily synthesized by one skilled in the art from procedures described herein, as well as in patents or patent applications listed herein which are all hereby incorporated by reference in their entireties and for all purposes as if fully set forth herein.
  • Compounds of the embodiments may generally be prepared using a number of methods familiar to one of skill in the art, such as, for example, the methods disclosed in the literature publications " Imidazo[1,2-a]quinoxalines: synthesis and cyclic nucleotide phosphodiesterase inhibitory activity.” Stephanie Parra, et. al. European Journal of Medicinal Chemistry, 2001, 36, pp. 255-264 ; " Design, synthesis and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine Al receptor antagonists.” Chun-He Liu, et. al. Bioorganic and Medicinal Chemistry, 2004, 12, pp.
  • Schemes 1-2 illustrate general methods for the preparation of intermediates and compounds of the embodiments. These compounds are prepared from starting materials either known in the art for commercially available. The specific compounds are for illustrative purposes only.
  • 2,3-dichloropyrazines ( 1.1 ), or equivalent reagents reacts with a substituted or unsubstituted oc-hydroxyalkylamine ( 1.2 ) to give a intermediates of Formula 1.3.
  • Oxidation of the alcohol of intermediates 1.3 under conditions known to those familiar in the art as a Swern oxidation, or equivalents, provides the aldehyde or ketone intermediates of Formula 1.4.
  • Treatment with a dehydrating reagent such as, for example, TFAA in a solvent such as, for example, TFA then provides the intermediates of Formula 1.5.
  • Treatment of intermediate 1.5 with ammonia then provides compounds of Formula 1.6.
  • intermediate 1.5 can be treated with reagents such as hydrazine, hydroxyamine, sodium azide or equivalents to provide intermediates of Formula 1.7. Treatment with under reducing conditions such as, for example, hydrogen with palladium on carbon in a solvent such as methanol or tetrahydrofuran then provides compounds of Formula 1.6. Alternately, intermediate 1.5 can be treated with a suitably protected amine to provide intermediates of Formula 1.8. Removal of the protecting groups provides compounds of the Formula 1.6.
  • reagents such as hydrazine, hydroxyamine, sodium azide or equivalents
  • Treatment with under reducing conditions such as, for example, hydrogen with palladium on carbon in a solvent such as methanol or tetrahydrofuran then provides compounds of Formula 1.6.
  • intermediate 1.5 can be treated with a suitably protected amine to provide intermediates of Formula 1.8. Removal of the protecting groups provides compounds of the Formula 1.6.
  • the 2.3-dichlorpyrazines of Formula 1.1 can be obtained commercially or by one trained in the art following chemistries similar to those found in " Product class 14: pyrazines.” N. Sato. Science of Synthesis 2004, 16, pp. 751-844 ; " Pyrazines. I. Syntheses of 2,3-dihydroxypyrazines and their derivatives.” Jiro Adachi, Nobuhiro Sato. Journal of Organic Chemistry 1972, 37(2), pp. 221-5 ; " Displacements and nuclear substitutions on hydroxypyrazines.” Geo Karmas. Paul E, Spoerri. Journal of the American Chemical Society 1956, 78, pp. 4071-7 ; each of which is incorporated by referene herein in its entiety for all purposes.
  • Scheme 2 depicts alternate methods for the preparation of compounds of the embodiment through an intermediate of Formula 2.1 prepared by methods described in Scheme 1.
  • Treatment of intermediates of the Formula 2.1 with a halogenating agent such as, for example, NBS provides the halogenated intermediate 2.2 which can be treated with methods, known to one trained in the art, for the removal of protecting groups to provide compounds of the embodiment of Formula 2.3.
  • Other halogenating agents can be used to in the conversion of intermediates of Formula 2.1 to 2.2.
  • Other halogenating agents include, but are not limited to, NIS, NCS, chlorine, bromine, iodine and ICl.
  • Deprotection of a compound of Formula 2.2 where P and P' are 4-methoxybenzyl groups treatment with an acid such as, for example, HCl in a solvent such as, for example, dioxane or THF can provide compounds of the embodiment of Formula 2.3.
  • an intermediate of Formula 2.2 where P and P' are taken together to form a phthalamide, or equivalent protecting group can be treated with a reagent such as, for example, hydrazine in a solvent such as, for example, methanol, tetrahydrofuran or dimethylformamide can provide compounds of the embodiment of Formula 2.3.
  • Treatment of compounds of Formula 2.1 with a strong base such as, for example, butyllithium in a solvent such as tetrahydrofuran followed by introduction of an electrophile such as, for example, methyl iodide or propylene oxide provides intermediates of the Formula 2.4.
  • a strong base such as, for example, butyllithium in a solvent such as tetrahydrofuran followed by introduction of an electrophile such as, for example, methyl iodide or propylene oxide
  • an electrophile such as, for example, methyl iodide or propylene oxide
  • Intermeidates of the Formula 3.5 can be treated with a suitably protected amine such as, for example, bis(4-methoxybenzyl)amine to provide intermediates of Formula 3.6.
  • a suitably protected amine such as, for example, bis(4-methoxybenzyl)amine
  • Removal of the 4-methoxybenzyl protecting groups with a reagents such as, for example, TFA provides compounds of the embodiment of Formula 3.7.
  • intermediates of the formual 3.6 can be treated with a strong base such as, for example, butyllithium in a solvent such as, for example, tetrahydrofuran followed by the addition of an electrophile such as, for example, isopropylene oxide to provide intermediates of the formua 3.8.
  • Removal of the 4-methoxybenzyl protecting groups with a reagents such as, for example, TFA provides compounds of the embodiment of Formula 3.9. The detailed preparaiton of these compounds is shown in Examples 3 and 4, below.
  • the title compound was synthesized generally according to the procedures described in Carine Deleuze-Masquefa, Grégori Gerebtzoff, Guy Subra, Jean-Roch Fabreguettes , Annabel Ovens, Ma ⁇ lle Carraz, Marie-Paule Strub, Jacques Bompart. Design and synthesis of novel imidazo[1,2-a]quinoxalines as PDE4 inhibitor. Bioorganic & Medicinal Chemistry (2004), 12(5), 1129-1139 . and in Chun-He Liu, Bo Wang, Wei-Zhang Li, Liu-Hong Yun, Ying Liu, Rui-Bing Su, Jin Li and He Liu.
  • the title compound was synthesized generally according to the procedures described in Chun-He Liu, Bo Wang, Wei-Zhang Li, Liu-Hong Yun, Ying Liu, Rui-Bing Su, Jin Li and He Liu. Design, synthesis, and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine A1 receptor antagonist. Bioorganic & Medicinal Chemistry (2004), 12(17), 4701-4707 .
  • the compounds of Examples 3 and 4 exhibited an EC 50 value with respect to agonism of IL-6 and TNF- ⁇ at less than 15 ⁇ M.
  • the compounds of Examples 3 and 4 were tested for stimulatory activity of human TLR 7 and 8 receptors and the mouse TLR7 receptor. Both compounds stimulated TLR7 and TLR8 activity, wherein the compound of Example 3 exhibited slightly more reactivity for mouse TLR7 than did the compound of Example 4.
  • the compound of Example 3 exhibited stimulatory activity with human TLR8 while the compound fo Example 4 exhibited little or no reactivity with human TLRB.
  • the compound of Example 3, and to a greater extent, Example 4 exhibited activity for stimulation of human TLR7.
  • each of these compounds is individually preferred and is preferred as a member of a group that includes any or all of the other compounds of Formula (I), or (II), or both.
  • Each compound is preferred in methods of modulating an immune response and in methods of treating biological conditions associated therewith, for example to be used as a vaccine adjuvant.
  • Each of the compounds is also preferred for use in preparation of medicaments for immunopotentiation, reducing tumor growth, treating microbial and viral infections, particularly influenza, HCV and HSV, and in treating biological conditions mediated therefrom.
  • Compounds found to not be effective at a concentration of 20 ⁇ M or less using the assay described below are also useful within the scope of the invention, since the invention is not meant to be limited to those compounds that are useful at a concentration of 20 ⁇ M or less.
  • Compounds may be useful as intermediates, or as final products that cause production of TNF- ⁇ at higher concentrations, such as 100 ⁇ M, 200 ⁇ M or 300 ⁇ M in the assays described herein.
  • Loxoribine causes useful production of TNF- ⁇ at 300 ⁇ M (see Pope et al. Cellular Immunology 162: 333-339 (1995 )).
  • Candidate small molecule immunopotentiators can be identified in vitro. Compounds are screened in vitro for their ability to activate immune cells. One marker of such activation is the induction of cytokine production, for example TNF- ⁇ production. Apoptosis inducing small molecules may be identified having this activity. These small molecule immuno-potentiators have potential utility as adjuvants and immuno-therapeutics.
  • HTS High Throughput Screening
  • SMIPs imidazoquinoxaline small molecule immune potentiators
  • PBMC peripheral blood mononuclear cells
  • FCS fetal calf serum
  • the robustness of the assay and low backgrounds allow for the routine selection of hits with ⁇ 10% of LPS activity that is normally between 5-10X background (cells alone). Selected hits are then subjected to confirmation for their ability to induce cytokines from multiple donors at decreasing concentrations. Those compounds with consistent activity at or below 5 ⁇ M are considered confirmed for the purposes of this assay.
  • the assay is readily modified for screening for compounds effective at higher or lower concentrations.
  • cytokines e.g., ILl-beta, IL-12, IL-6, IFN-gamma, IL-10 etc.
  • cytokines e.g., ILl-beta, IL-12, IL-6, IFN-gamma, IL-10 etc.
  • SMIP or composition comprising a SMIP of the preferred embodiments of the present invention can be implemented using methods known in the art, such as by measuring antigen specific antibody production, activation of specific populations of lymphocytes such as CD4 + , CD8+ T cells or NK cells, and/or production of cytokines such as IFN, IL-2, IL-4 or IL-12.
  • Methods for measuring specific antibody responses include enzyme-linked immunosorbent assay (ELISA) as known in the art.
  • Measurement of numbers of specific types of lymphocytes such as CD4 + T cells can be achieved, for example, with fluorescence-activated cell sorting (FACS).
  • Cytotoxicity assays can also be performed using methods known in the art, e.g., as described in Raz et al., (1994) Proc. Natl. Acad. Sci. USA 91:9519-9523 . Serum concentrations of cytokines can be measured, for example, by ELISA. Such assays are described, e.g., in Selected Methods in Cellular Immunology (1980) Mishell and Shiigi, eds., W.H. Freeman and Co .
  • Human blood from one or multiple human donors were collected into the BD VacutainerTM CPT tube with sodium citrate (BD, Franklin Lakes, NJ), and spun for 20 minutes at 1600g. After centrifugation, mononuclear cells in the top layer in the tubes were collected and then washed three times with PBS buffer. The washed cells were then reconstituted at a required cell concentration in complete RPMI containing 10% FBS plus 100 units/ml penicillin and 100ug/ml streptomycin.
  • Spleens were isolated from Balbc mice and minced to release the splenocytes from the tissues. After the minced samples were treated ammonium salt to destroy the red blood cells, the rest of the spleenocytes were washed and reconstituted at a required cell concentration with completed RPMI medium.
  • the human myelomonocytic transformed cell line is responsive to TLR8 agonists and weakly to TLR7 agonists.
  • the cell line is cultured in RPMI medium supplemented with 10% FBS.
  • hPBMC Human PBMC
  • mouse spleen cells at 5 million cells/ml
  • human monocytic THP-1 cells at 1 million cells/ml
  • tested compounds such as imidazoquinoxalines at titrated compound concentrations in the complete RPMI medium.
  • the culture supernatant was collected and assayed for the secreted cytokines in the presence of the compounds.
  • Human or mouse Beadlyte multi-cytokine flex kits (Upstate, Lake Placid, NY) were used to measure the amount of the following cytokines: TNF- ⁇ and IL-6 according to the manufacturers instructions.
  • HEK293 cells (ATCC, CRL-1573) are seeded in a T75 flask at 3x10 6 in 20ml of DMEM supplemented with 0.1mM nonessential amino acid, 1mM sodium pyruvate, 2mM L-glutamine, penicillin-streptomycin, and 10% FCS.
  • the cells are transfected with 1) pNFkB-TA-luciferase reporter (0.4ug) (BD clontech, Palo Alto, CA), and with 2) with pGL4.74 (0.01ug) that carries a TK promoter, not responsive to NF-kB stimulation, and carries a Renilla luciferase gene, used as an internal control (Promega, WI), and 3), separately with a following TLR construct (10 ug): human TLR (hTLR) 7, hTLR8, mouse TLR7 (mTLR7) puno constructs (Invivogene, CA), using Fugene 6 transfection reagent (Roche).
  • TLR construct 10 ug): human TLR (hTLR) 7, hTLR8, mouse TLR7 (mTLR7) puno constructs (Invivogene, CA), using Fugene 6 transfection reagent (Roche).
  • the transfected cells after 24 hours transfection are collected and seeded in a 96-well and flat-bottom plate (1x10 4 cell/well) plate, and stimulated with the test compounds at the following concentrations: 30, 10, 3, 1, 0.3, 0.1, 0.03 uM. After overnight compound stimulation, the cells are assayed for expression of fly and renilla luciferases using Dual-Luciferase Reporter Assay System (Promega, WI). NF-kb activation is directly proportional to relative fly luciferase units, which is measured against the internal control renilla luciferase units.
  • compound ranking is based on potency in cell-based screens for cytokine induction. Briefly, the EC 50 of each compound for a given cytokine is calculated relative to a reference composition (i.e. LPS or other potent immune stimulator). This value is then used as the divisor of the maximum level of cytokine produced (pg/ml) in the assay. Five parameter curve fitting of cytokine dose response curves to different SMIPs for the indicated cell populations is used to calculate EC50. Rank-scoring of SMIP potency is calculated by dividing the maximum concentration of cytokine produced by the relative EC50 established for each compound indicated.
  • a reference composition i.e. LPS or other potent immune stimulator
  • gp120dV2EnvSF162 antigen recombinant gp120 protein derived from sequence of HIV-1 strain SF162 - the V2 domain is deleted; Pharm Res. 2004 Dec 21(12):2148-52 ) as mixed with 50 microliters of MF59 adjuvant, followed the by the addition of 0, 1, 5, or 25 micrograms of a small molecule immune potentiator (SMIP) and adjusted to 100 microliters with PBS. 50 microliters of the solution is subsequently injected into both the left and right tibialis anterior muscles of female BALB/c mice (Day 0), for a total volume of 100 microliters per mouse.
  • SMIP small molecule immune potentiator

Abstract

The invention provides novel compositions comprising imidazoquinoxaline compounds of formula (I). Also provided are the compounds of formula (I) for use in methods of administering the compositions in an effective amount to enhance the immune response of a subject. Further provided are novel compositions and the compositions for use in methods of administering the compositions in combination with (an) other agent(s).

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Application Serial No. 60/785,545, filed on March 23, 2006 , which is hereby incorporated by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention generally relates to small molecule immune potentiators (SMIPs) that are novel imidazoquinoxaline compounds and analogs thereof that are capable of stimulating or modulating an immune response in a subject. The invention also relates to novel combinations of antigens with the immune potentiators that may be used in vaccine therapies. In some embodiments, the compounds can be used as immunotherapeutic agents for proliferative diseases, infectious diseases, autoimmune diseases, allergies, and/or asthma.
  • BACKGROUND OF THE INVENTION
  • Issued U.S. Patent Nos. 4,689,338 , 5,389,640 , 5,268,376 , 4,929,624 , 5,266,575 , 5,352,784 , 5,494,916 , 5,482,936 , 5,346,905 , 5,395,937 , 5,238,944 , 5,525,612 , and 6,110,929 , and WO 99/29693 disclose imidazoquinoline compounds of the general structure (a) for use as "immune response modifiers":
    Figure imgb0001
    Each of these references is hereby incorporated by reference in its entirety and for all purposes as if fully set forth herein.
  • U.S. Patent No. 6,083,505 , describes specific imidazoquinolines for use as adjuvants. WO 03/097641 discloses the use of certain imidazoquinolines and salts thereof for the treatment of certain protein kinase dependent diseases and for the manufacture of pharmaceutical preparations for the treatment of diseases.
  • Immune response to certain antigens can be enhanced through the use of immune potentiators, known as vaccine adjuvants. Such adjuvants potentiate the immune response to specific antigens and are, therefore, the subject of considerable interest and study within the medical community.
  • Research has resulted in the development of vaccines possessing antigenic epitopes that were previously impossible to produce. For example, currently available vaccine candidates include synthetic peptides mimicking numerous bacterial and viral antigens. The immune response to these purified antigens can be enhanced by coadministration of an adjuvant. Unfortunately, conventional vaccine adjuvants possess a number of drawbacks that limit their overall use and effectiveness. Moreover, many of the adjuvants currently available have limited utility because they include components that are not metabolized by humans. Additionally, most adjuvants arc difficult to prepare and may require time-consuming procedures and, in some cases, the use of elaborate and expensive equipment to formulate a vaccine and adjuvant system.
  • Immunological adjuvants are described in "Current Status of Immunological Adjuvants", Ann. Rev. Immunol., 1986, 4, pp. 369-388, and "Recent Advances in Vaccine Adjuvants and Delivery Systems" by Derek T O'Hagan and Nicholas M. Valiante. See also U.S. Patent Nos. 4,806,352 ; 5,026,543 ; and 5,026,546 for disclosures of various vaccine adjuvants appearing in the patent literature. Each of these references is hereby incorporated by reference in its entirety and for all purposes as if fully set forth herein.
  • Efforts have been made to identify new immune modulators for use as adjuvants for vaccines and immunotherapies that would overcome the drawbacks and deficiencies of conventional immune modulators. In particular, an adjuvant formulation that elicits potent cell-mediated and humoral immune responses to a wide range of antigens in humans and domestic animals, but lacking the side effects of conventional adjuvants and other immune modulators, would be highly desirable. This need could be met by small molecule immune potentiators (SMIPs) because the small molecule platform provides diverse compounds for the selective manipulation of the immune response, necessary for increasing the therapeutic index immune modulators.
  • Novel sole-acting agents with varied capacities for altering levels and/or profiles of cytokine production in human immune cells are needed. Compounds with structural disparities will often elicit a desired response through a different mechanism of action, or with greater specificity to a target, such as a dendritic cell, modulating potency and lowering side effects when administered to a patient.
  • The immunosuppressive effect of cytostatic substances has rendered them useful in the therapy of autoimmune diseases such as multiple sclerosis, psoriasis and certain rheumatic diseases. Unfortunately, their beneficial effect has to be weighed against serious side effects that necessitate dosages that are too low. Furthermore, interruption of the treatment may be required.
  • Agents and/or combinations of active substances that result in significantly improved cytostatic or cytotoxic effects compared to conventional cytostatics, e.g., vincristin, methotrexate, cisplatin, etc., are needed. With such agents and combinations, chemotherapies may be offered that combine increasing efficiency with a large reduction of side effects and therapeutic doses. Such agents and combination therapies may thus increase the therapeutic efficiency of known cytostatic drugs. In some embodiments, the compounds of the invention are used in combination with compounds that provide significantly improved cytostatic or cytotoxic effect compared to conventional cytostatic agents when administered alone. Additionally, cell lines that are insensitive to conventional chemotherapeutic treatment may also be susceptible to chemotherapy using combinations of active substances.
  • Improved methods for preparing therapeutics that serve to augment natural host defenses against viral and bacterial infections, or against tumor induction and progression, with reduced cytotoxicity, are needed. The present invention provides such methods, and further provides other related advantages. The current invention provides method of preparing therapeutic and prophylactic agents for treatment of disease states characterized by other immune deficiencies, abnormalities, or infections including autoimmune diseases and viral and bacterial infections responsive to compounds with the capacity to modulate cytokines and/or TNF-α.
  • BRIEF SUMMARY OF THE INVENTION
  • The instant invention provides novel immune potentiators, immunogenic compositions, novel compounds and pharmaceutical compositions, and novel methods of administering a vaccine, by administering small molecule immune potentiators alone or in combination with antigens and/or other agents. The invention further provides novel compounds and pharmaceutical compositions, for use in the treatment of cancer, precancerous lesions, autoimmune diseases, infectious diseases, allergies, and asthma. The invention further provides the use of the compounds of the invention in the manufacture of medicaments for use in the treatment of cancer, precancerous lesion, autoimmune diseases, allergies, and asthma.
  • The imidazoquinoxaline compounds and analogs thereof used in the methods and compositions of the invention are inexpensive to produce and easy to administer. They have potential for finer specificity compared to existing immunostimulants, thus providing improved efficacy and safety profiles.
  • As adjuvants, the imidazoquinoxaline compounds and analogs thereof may be combined with numerous antigens and delivery systems to form an immunogenic composition. In a preferred embodiment, the immunogenic composition can be used in the manufacture of a vaccine or a medicament.
  • As immunotherapeutics, the imidazoquinoxaline compounds and analogs thereof are used alone or in combination with other therapies (e.g., anti-virals, anti-bacterials, other immune modulators or in therapeutic vaccine antigens) for treatment of the following: persistent of chronic viral infections such as, e.g., those caused by the human immunodeficiency virus (HIV), the hepatitis C virus (HCV), the hepatitis B virus (HBV), the herpes simplex virus (HSV); persistent or chronic bacterial infections, such as those caused by Chlamydia, pseudomonas, gonorrhea, treponema pallidium (syphilis), H. pylori, tuberculosis, Lyme disease; chronic or persistent fungal infections, chronic or persistent parasitic infections (e.g., malaria); as well as medicaments for the reduction of tumor growth or modulation of abnormal cellular proliferation associated with diseases such as actinic kcratosis, atypical or dysplastic ncvi, or premalignant lentigos.
  • The imidazoquinoxaline compounds and analogs thereof of the present invention may target substrates in the disease state, such as, for example particular kinases including EGFr, c-Kit, bFGF, Kdr, CHK1, CDK, cdc-2, Akt, PDGF, PI3K, VEGF, PKA, PKB, src, c-Met, Abl, Ras, RAF, and MEK, among others.
  • As immunotherapeutics, the imidazoquinoxaline compounds and analogs thereof may also be used for the treatment of cancer either alone or in combination with other anticancer therapies (e.g., chemotherapeutic agents, (monoclonal antibodies) mAbs or other immune potentiators). In addition, certain imidazoquinoxalines with the capacity to induce Type 1 cytokines (e.g., IL-12, TNF-α or IFN's) may be used for the treatment of allergies and/or asthma due to their capacity to steer the immune response towards more benign sequelae. The imidazoquinoxaline compounds and analogs thereof may be used, for example, for the treatment of bacillus Calmette-Guerin (BCG), cholera, plague, typhoid, hepatitis B infection, influenza, inactivated polio, rabies, measles, mumps, rubella, oral polio, yellow fever, tetanus, diphtheria, hemophilus influenza b, meningococcus infection, and pneumococcus infection. The imidazoquinoxaline compounds and analogs thereof may be used in an anti cell proliferative effective amount for the treatment of cancer. The imidazoxaquinoline compounds may also be used in anti-Th2/Type2 cytokine amount for the deviation of allergic/asthmatic immune responses.
  • In some embodiments, methods of treating cancer and/or precancerous lesions are provided. In such embodiments, one or more known anticancer agent is combined with one or more imidazoquinoxaline compound to reduce tumor growth in a subject. A number of suitable anticancer agents are contemplated for use in the methods of the present invention and are described more thoroughly in the following detailed description.
  • In accordance with another embodiment, there is provided a method of inhibiting tumor cell growth in a subject. The method includes administering to a subject an effective dose of a combination comprising at least one imidazoquinoxaline compound as described herein, and a monoclonal antibody (mAb). The combination may be more effective at inhibiting such cell growth than when the mAb is administered by itself. In some embodiments of the methods of treating cancer with the combination, an additional imidazoquinoxaline compound as described herein compound and/or mAb, is administered to the subject.
  • In some embodiments, the invention provides immunogenic compositions comprising an antigen and an imidazo[1,2-a]quinoxalin-4-amine effective to stimulate a cell mediated response to said antigen. In some embodiments, the imidazo[1,2-a]quinoxalin-4-amine compounds have the general Formula described herein. Accordingly, in some embodiments of the methods and compositions of the invention, the imidazoquinoxaline compound has the Formula (I):
    Figure imgb0002
    wherein:
    • R1 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, - S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • R6 is selected from the group consisting of hydrogen, halogen, hydroxy, -O-C1-3 alkyl, C1-3 haloalkyl, -O-C1-3 haloalkyl, C1-3 perhaloalkyl, -O-C1-3 perhaloalkyl, C1-3 hydroxyalkyl, -O-C1-3 hydroxyalkyl, CN, -(CH2)qC(=O)R7, -O-(CH2)qC(=O)R7, -(CH2)qN(R8)(R9), -S-C1-3 alkyl, -S(=O)2-R10 and -S(=O)2N(R8)(R9);
    • q is 0, 1, 2 or 3;
    • R7 is selected from hydrogen, hydroxy, C1-3 alkyl or C1-3 alkoxy; and
    • R8, R9 and R10 are each independently hydrogen or C1-3 alkyl;
    • provided that R1, R2 and R6 are not simultaneously hydrogen; and
    • if R1 and R6 are both H, R2 is not butyl;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In other embodiments of the methods and compositions of the invention, the compound has the Formula (II):
    Figure imgb0003
    wherein:
    • R1 is selected from the group consisting of hydroxy, amino, -OR3, -N(R3)(R4), -NR3C(-O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -S(=O)pR4, and S(=O)pNR3R4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, - S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • Ra and Rb are independently selected from the group consisting of hydrogen, CN, NO2, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • q is 0, 1, 2 or 3;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In other embodiments of the methods and compositions of the invention, the compound has the Formula (II):
    Figure imgb0004
    wherein:
    • R1 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • Ra and Rb are taken together to form a group selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocyclyl, or substituted heterocyclyl, with the proviso that Ra and Rb are not taken together to form a thiophenyl group;
    • q is 0, 1, 2 or 3;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • Methods of manufacturing compounds and compositions described herein are provided and contemplated to fall within the scope of the invention as is the use of the imidazoquinoxalines in methods for manufacturing medicaments for use in the methods of the invention.
  • In each of the embodiments of the invention, compounds of Formula (I) or (II), can be used in the manufacture of a medicament for enhancing the immune response to an antigen.
  • Other embodiments provide the use of the compounds of the invention, in the manufacture of medicament for immune stimulation, and another agent, such as an antigen, for simultaneous separate or sequential administration. In another more particular embodiment the use is for treating or preventing a bacterial or viral infection. In another embodiment the use is for treating cancer. In another embodiment the use is for preventing influenza infection.
  • Other embodiments provide a pharmaceutical preparation or system, comprising (a) a compound of Formula (I) or (II); and (b) an antigen, wherein (a) and (b) are either in admixture or arc separate compositions. The agents arc for simultaneous separate or sequential administration. In another more particular embodiment the use is for preventing a viral, bacterial, fungal or parasitic infection. In another embodiment the use is for treating cancer.
  • Further embodiments of the invention include those described in the detailed description.
  • DETAILED DESCRIPTION OF THE DRAWINGS
    • FIG. 1 illustrates stimulatory activity of compounds described in Examples 3 and 4 was tested against TLR 7 and 8 receptors and the mouse TLR7 receptor.
    • FIG. 2 illustrates human PBMC (peripheral blood mononuclear cell) TNF-α production as a function of the concentration of the compounds of Examples 3 and 4.
    DETAILED DESCRIPTION OF THE INVENTION
  • Applicants have discovered methods of stimulating cytokine activity in cells and immunotherapeutics and/or vaccine adjuvants, that will provide effective treatments for disorders such as those described herein and those apparent to one skilled in the art.
  • In one embodiment, the invention provides a compound of Formula (I):
    Figure imgb0005
    wherein:
    • R1 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, - NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, - NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R-4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • R6 is selected from the group consisting of hydrogen, halogen, hydroxy, -OR3, - N(R3)(R4), C1-3 haloalkyl, -O-C1-3 haloalkyl, C1-3 perhaloalkyl, -O-C1-3 perhaloalkyl, C1-3 hydroxyalkyl, -O-C1-3 hydroxyalkyl, CN, -(CH2)qC(=O)R7, -O-(CH2)qC(=O)R7, - (CH2)qN(R8)(R9), -S-C1-3 alkyl, -S(=O)2-R10 and -S(=O)2N(R8)(R9);
    • q is 0, 1, 2 or 3;
    • R7 is selected from hydrogen, hydroxy, C1-3 alkyl or C1-3 alkoxy; and
    • R8, R9 and R10 are each independently hydrogen or C1-3 alkyl;
    • provided that R1, R2 and R6 are not simultaneously hydrogen; and
    • if R1 and R6 are both H, R2 is not butyl;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In one preferred embodiment, if R2 and R6 are H, then R1 is not methyl.
  • In one preferred embodiment, if R2 and R6 are H, then R1 is not isobutyl.
  • In one preferred embodiment, if R2 and R6 are H, then R1 is not phenethyl.
  • In some embodiments, R1 is C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, or substituted C2-C12 alkenyl. In some embodiments, R1 is hydrogen, C1-C12 alkyl or substituted C1-C12 alkyl. In some further embodiments, R1 is C1-C12 alkyl. In some further embodiments, R1 is C1-C6 alkyl, for example isobutyl (i.e., -CH2-CH(CH3)2).
  • In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen or hydroxyalkyl, for example -CH2-C(OH)(CH3)2.
  • In some embodiments, R1 is C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, or substituted C2-C12 alkenyl; and R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl. In some embodiments, R1 is C1-C12 alkyl or substituted C1-C12 alkyl; and R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl. In some embodiments, R1 is C1-C12 alkyl; and R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl. In some embodiments, R1 is C1-C6 alkyl; and R2 is hydrogen or hydroxyalkyl. In some embodiments, R1 is -CH2-CH(CH3)2; and R2 is hydrogen or hydroxyalkyl, for example -CH2-C(OH)(CH3)2.
  • In some embodiments, R6 is -OR3 or -N(R3)(R4). In other embodiments, only one of R1, R2 and R6 is H.
  • In some of each of the foregoing embodiments, R6 is hydrogen. In some embodiments, the compound is selected from
    Figure imgb0006
    and pharmaceutically acceptable salts, tautomers and salts of the tautomers thereof.
  • In other embodiments, the compound is selected from the group consisting of:
    Figure imgb0007
    Figure imgb0008
    Figure imgb0009
    Figure imgb0010
    Figure imgb0011
    Figure imgb0012
    Figure imgb0013
  • In other embodiments of the methods and compositions of the invention, the compound has the Formula (II):
    Figure imgb0014
    wherein:
    • R1 is selected from the group consisting of hydroxy, amino, -OR3, -N(R3)(R4), - NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -S(=O)pR4, and S(=O)pNR3R4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, - NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • Ra and Rb are independently selected from the group consisting of hydrogen, CN, NO2, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), - NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, - S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • q is 0, 1, 2 or 3;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In another more particular embodiment R1 is -N(R3)(R4). More particular still, R3 and R4 arc each independently selected from the group consisting of hydrogen, C1-C12 alkyl, and substituted C1-C12 alkyl. In another embodiment R1 is hydroxy.
  • In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen or hydroxyalkyl, for example -CH2-C(OH)(CH3)2.
  • In other embodiments of Formula (II):
    • R1 is selected from the group consisting of C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted G2-G12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, - C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, - NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • Ra and Rb are independently selected from the group consisting of hydroxy, amino, - OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, - S(=O)pR4;
    • q is 0, 1, 2 or 3;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In other embodiments of the methods and compositions of the invention, the compound has the Formula (II):
    Figure imgb0015
    wherein:
    • R1 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, - NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino, -OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, - NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
    • Ra and Rb are taken together to form a group selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocyclyl, or substituted heterocyclyl, with the proviso that Ra and Rb are not taken together to form a thiophenyl group;
    • q is 0, 1, 2 or 3;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a
    pharmaceutically acceptable salt of the tautomer.
  • In a more particular embodiment, Ra and Rb are taken together to form a 6-membered substituted or unsubstituted heteroaryl group, more particularly a substituted or unsubstituted pyridyl group.
  • In a more particular embodiment, Ra and Rb are taken together to form a saturated group selected from a heterocyclyl, substituted heterocyclyl, cycloalkyl or substituted cycloalkyl.
  • In a more particular embodiment, Ra and Rb are taken together to form a substituted or unsubstituted aryl group, more particularly a substituted or unsubstituted phenyl group.
  • In some embodiments, R1 is C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, or substituted C2-C12 alkenyl; and R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl. In some embodiments, R1 is C1-C12 alkyl or substituted C1-C12 alkyl; and R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl. In some embodiments, R1 is C1-C12 alkyl; and R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl. In some embodiments, R1 is C1-C6 alkyl; and R2 is hydrogen or hydroxyalkyl. In some embodiments, R1 is -CH2-CH(CH3)2; and R2 is hydrogen or hydroxyalkyl, for example -CH2-C(OH)(CH3)2.
  • In another more particular embodiment R1 is -N(R3)(R4). More particular still, R3 and R4 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, and substituted C1-C12 alkyl. In another embodiment R1 is hydroxy.
  • In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl. In some embodiments, R2 is hydrogen or hydroxyalkyl, for example -CH2-C(OH)(CH3)2.
  • In some embodiments of the compounds having the Formula II, wherein Ra and Rb together form an unsubstituted phenyl group; R1 and R2 are not simultaneously hydrogen, and if R2 is hydrogen, then R1 is not methyl, isobutyl, or phenethyl.
  • Further provided are compounds of Formula (I) or (II) and mixtures thereof where any asymmetric carbon atom(s) can have either the R or S configuration. Substituents at a double bond or a ring of the compounds of Formula (I) or (II) may be present in either the cis (-Z-) or trans (-E-) configurations. The compounds may thus be present as mixtures of isomers, diastereomers, and enantiomers or may be present as pure isomers. In some embodiments, the compounds are enantiomerically pure where only one enantiomer is present. In other embodiments, the compound may be present as a mixture of enantiomers which includes more of one enantiomer than it does of the other.
  • Generally, a compound of the invention, or a composition comprising such a compound, is considered effective to elicit an immune response at a concentration of 300 µM or less in some embodiments, 200 µM or less in some embodiments, 100 µM or less in some embodiments, or 20 µM or less in some embodiments if the compound of the invention effects (a) the production of TNF-α in an in vitro cell based assay of human peripheral blood mononuclear cells, and (b) a concentration of human peripheral blood mononuclear cells (PBMCs) of about 500,000/mL, when the cells are exposed to the compound for a period of about 18-24 hours, preferably about 24 hours.
  • The above method of stimulating a local immune response, for example in selected cells or tissues of a patient, includes the stimulation of a local immune response where the selected cells or tissues are infected or cancerous. In some embodiments, the selected cells or tissues are infected with a fungus or bacterium. In some embodiments, the selected tissues are inflamed with an allergen, for example in an asthmatic condition. In other embodiments, the selected cells are infected with a virus or bacteria. In still other embodiments, the infectious agent is HCV, HIV, HBV, HSV, H. pylori, HSV Type 1 or 2, or Human Papilloma Virus.
  • Another embodiment provides a method of inducing interferon biosynthesis in a subject. Such methods include administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce interferon biosynthesis. In some such methods, a vaccine adjuvant of Formula (I) or (II) is administered to the subject in an amount sufficient to induce interferon biosynthesis.
  • Another embodiment provides a compound of Formula (I) or (II), wherein the compound is co-administered with another agent to a patient in need thereof. In some such embodiments, the agent is an antigen or a vaccine. In embodiments, where the compound of Formula (I) or (II) is co-administered to a patient or subject along with another agent, the compound of Formula (I) or (II) may be administered to the subject before, during, or after the other agent is administered to the subj ect. Therefore, in some embodiments, the compound of Formula (I) or (II) is administered to the subject at the same time that the other agent is administered to the subject. The location or site of administration of the compound of Formula (I) or (II) can be the same or different as the location of an antigen when the compound is used with an antigen.
  • Another embodiment provides a method of modulating an immune response in a subject. Such methods include administering a compound of Formula (I) or (II) to the subject.
  • Another embodiment provides a method for inducing the production of TNF-α in a subject. Such methods include administering a compound of Formula (I) or (II) to a subject in an amount sufficient to induce the production of TNF-α. In some such embodiment thereof, the compound has an average steady state drug concentration in the blood of less than 20 µM.
  • Another embodiment provides a method of inducing an immune response in a subject. The embodiment includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response. In some such embodiments, the immune response includes the production of cytokines or increased production of TNF-α. In some embodiments, the induction of an immune response includes the production of antibodies that may be neutralizing antibodies or antibodies that mediate Antibody Dependent Cell Mediated Cytotoxicity (ADCC antibodies).
  • Another embodiment provides a method of inducing an immune response in a subject suffering from a microbial (viral, bacterial, fungal or parasitic) infection. The method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from a viral infection or a disease condition caused by a virus. The method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response in the subject. The virus may be selected from one or more of the viral pathogens described in the antigen section below. In some such embodiments, the subject is suffering from a viral infection or disease condition caused by the hepatitis C virus (HCV). In other embodiments, the subject is suffering from a viral infection or disease condition caused by the human immunodeficiency virus (HIV).
  • In other embodiments, the immune response is induced un a subject suffering from a bacterial, fungal or parasitic infection wherein the disease causing organism may be selected from one or more of the bacterial, fungal or parasitic pathogens described in the antigen section below.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from an abnormal cellular proliferation or cancer. The method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response. In some embodiments, the compound is administered to a subject that is suffering from a disease associated with abnormal cellular proliferation. In some such embodiments, the disease is selected from neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogenesis, or endotoxic shock.
  • Other embodiments provide methods of inducing an immune response in a subject suffering from an allergic disease. Such methods include administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response.
  • Another embodiment provides a method of inducing an immune response in a subject suffering from asthma. The method includes administering a compound of Formula (I) or (II) to the subj ect in an amount sufficient to induce an immune response. In some embodiments, asthma may be treated by steering the immune response away from Type 2 cytokine secretion and effector mechanism (e.g., IgE production and/or mast cell/basophil activation).
  • Another embodiment provides a method of inducing an immune response in a subject suffering from precancerous lesions. The method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to induce an immune response. In some such embodiments, the precancerous lesions are actinic keratosis. In other embodiments, the precancerous lesions are selected from actinic keratosis, atypical or dysplastic nevi, or premalignant lentigos. In another embodiment or method, the compound of Formula (I) or (II) is administered topically to a subject.
  • Other embodiments provide a method of inhibiting a kinase in a subject. Such methods include administering the compound of Formula (I) or (II) to the subject.
  • Another embodiment provides a method of modulating an immune response in a subject. The method includes administering a compound of Formula (I) or (II) to the subject in an amount sufficient to inhibit a kinase in the subject. In some such embodiments, the kinase is selected from EGFr, c-Kit, bFGF, Kdr, CHK1, CDK, cdc-2, Akt, PDGF, PI3K, VEGF, PKA, PKB, src, c-Met, Abl, Ras, RAF, MEK, or combinations thereof. In another embodiment or method, the compound of Formula (I) is administered topically to a subject.
  • Another embodiment provides a method of inducing an immune response in a subject, comprising: administering to the subject a compound of Formula (I) or (II) and an antigen, wherein the compound induces or enhances an immune response to the antigen in the subject. More particularly the antigen can be one or more viral, bacterial, fungal, parasitic or tumor antigens or other antigens as described herein.
  • Another embodiment provides a composition comprising: the compound of Formula (I) or (II) and another agent. In some embodiments, the other agent is an antigen. In further embodiments, the composition of the invention comprises the compound of Formula (I) or (II) with an antigen and a second adjuvant. In another embodiment, the composition of the invention provides a compound of Formula (I) or (II) and a second adjuvant. In another embodiment, the composition further comprises poly(lactide-co-glycolide) (PLG). In another embodiment, the composition further comprises MF59 or another adjuvant.
  • Additional embodiments, methods and compositions contemplated to be useful in the instant invention are disclosed in PCT/US2005/032721 , PCT/US2005/022769 , PCT,US2005/022520 and U.S.S.N. 10/814,480 , 10/762,873 , 60/582,654 , 10/405,495 , and 10/748,071 which are each hereby incorporated by reference in their entireties and for all purposes as if set forth fully herein.
  • Another embodiment provides a pharmaceutical composition, comprising: the compound of Formula (I) or (II) and a pharmaceutically acceptable excipient.
  • Another embodiment of the present invention provides a method of stimulating TLR-7 production comprising administering a compound of Formula (I), or (II) Another embodiment provides a method of stimulating TLR-8 production comprising administering a compound of Formula (I). Another embodiment provides a method of stimulating TLR-7 and TLR-8 production comprising administering a compound of Formula (I) or (II).
  • Compounds of the present invention cause immune potentiation and stimulate production of TLR-7 and TLR-8. Such compounds can be used as polyclonal activators for the production of antigens. More particularly the invention relates to a method of preparing monoclonal antibodies with a desired antigen specificity comprising contacting the compounds of the present invention (such as those of Formula (I) or (II)) with immortalized memory B cells.
  • The monoclonal antibodies produced therefrom, or fragments thereof may be used for the treatment of disease, for the prevention of disease or for the diagnosis of disease. Methods of diagnosis may include contacting an antibody or an antibody fragment with a sample. The methods of diagnosis may also include the detection of an antigen/antibody complex.
  • The memory B cells to be transformed can come from various sources (e.g. from whole blood, from peripheral blood mononuclear cells (PBMCs), from blood culture, from bone marrow, from organs, etc.), and suitable methods for obtaining human B cells are well known in the art. Samples may include cells that are not memory B cells or other blood cells. A specific human memory B lymphocyte subpopulation exhibiting a desired antigen specificity may be selected before the transformation step by using methods known in the art. In one embodiment, the human memory B lymphocyte subpopulation has specificity for a virus e.g. the B cells are taken from a patient who is suffering or has recovered from the virus. In another embodiment, B cells are taken from subjects with Alzheimer's disease and include B cells with specificity for B-amyloid (e.g. Mattson & Chan (2003) Science 301:1 847-9; etc.).
  • Another embodiment provides a method for producing immortalized B memory lymphocytes, comprising the step of transforming B memory lymphocytes using the Epstein Barr virus in the presence of a compound of the present invention, such as a compound of Formula (I) or (II). See WO 04/76677 .
  • The invention also provides pharmaceutical compositions that include any of the aforementioned compounds or embodiments of Formula (I) or (II). Such compositions may include other pharmaceutically acceptable ingredients such as one or more of excipients, carriers, and the like well-known to those skilled in the art.
  • It is contemplated that the invention encompasses all possible combinations of the preceding embodiments. In some embodiments of each of the compound and methods described herein, R6 of the compounds of Formula (I) is hydrogen.
  • The imidazoquinoxaline compounds and analogs thereof can be used with or without an antigen in therapeutic applications, for example to treat cancer or infectious diseases. The imidazoxaquinoline compounds may also be used in combination with other therapeutic agents, such as anti-viral agents and monoclonal antibodies in different therapeutic applications.
  • One embodiment of the method of inducing an immunostimulatory effect in a patient is directed to administering an immunogenic composition comprising an antigen in an amount effective to stimulate an immune response such as a cell-mediated immune response and, as a vaccine adjuvant, an imidazoquinoline compound, in an amount effective to potentiate the immune response such as the cell-mediated immune response to the antigen.
  • Agents combined with the imidazoquinoxaline compounds and analogs thereof, contemplated to be useful in treating the aforementioned diseases include those well known in the art, such as, but not limited to, anesthetics, hypnotic sedatives, anti-anxieties, antiepileptics, antipyretic antiphlogistics, stimulants, wake amines, anti-Parkinson drugs, agents for psychoneuroses, agents for central nervous system, skeletal muscle relaxants, agents for autonomic nervous system, antispastic agents, cytotoxic agents, monoclonal antibodies, drugs for eye, drugs for nose and ear, anti-vertiginous drugs, cardiotonics, antiarrhythmic drugs, diuretics, pressure reduction drugs, vasoconstrictors, coronary vasodilators, peripheral vasodilating drugs, hyper-lipemia drugs, breath stimulants, antitussive and expectorant drugs, bronchodilators, drugs for allergy, antidiarrheal drugs, drugs for intestinal disorders, peptic ulcer drugs, stomachic digestants, antacids, cholagogouses, pituitary hormone drugs, salivary gland hormones, thyroid hormone drugs, antithyroid drugs, anabolic steroids, corticosteroids, androgen drugs, estrogen drugs, corpus luteum hormone drugs, mixed hormones, urinary/genital organ drugs, anus drugs, surgical sterilizations/antiseptics, wound protectives, externals for purulent diseases, analgesics, antipruritics, astringents, antiphlogistics, externals for parasite skin diseases, skin-softening drugs, caustics, dental/oral drugs, vitamins, inorganic preparations, supplemental liquids, hemostatics, anticoagulation drugs, drugs for liver diseases, antidotes, habitual intoxication drugs, drugs for treatment of gout, enzyme preparations, diabetic drugs, antioncotics, antihistaminics, antibiotics (such as ketolides, aminoglycosides, sulphonamides, and/or beta lactams), chemotherapeutics, biological preparations, anthelmintics, anti-Protozoas, drugs for preparations, X-ray contrast media, and diagnostic drugs.
  • Further methods of the invention are provided wherein compositions described herein are used for the treatment of cancer and reduction of tumor growth. In one aspect, an imidazoxaquinoline compound of the invention is combined with a known mAb for the treatment of cancer. In one such embodiment, an antibody and an imidazoxaquinoline compound are administered to a subject in need thereof. In some such embodiments, the antibody, individually, has an inhibiting effect upon tumor cell growth, and the imidazoxaquinoline compound induces the production of cytokines.
  • In accordance with another embodiment of the present invention, a therapeutic composition for inhibiting tumor cell growth in a subject is provided. Such compositions include an effective amount of a combination of at least one imidazoquinoxaline compound of the invention, at least one mAb, and at least one pharmaceutically acceptable carrier. In such embodiments, the combination is expected to be more effective at inhibiting the growth of certain mammalian tumor cells than are any of the agents when individually administered.
  • In another embodiment, methods of treating cancer are provided in which known anticancer agents are combined with imidazoquinoxaline compounds and analogs thereof of the invention to reduce tumor growth in a subject. A number of suitable anticancer agents arc contemplated for use in such methods. Indeed, the present invention contemplates, but is not limited to, administration of numerous anticancer agents including, but not limited to: fcnrctinidc, vatalanib, SU-11248, SU 5416, SU 6668, oxaliplatin, bortczomib, R 115777, CEP-701, ZD-6474, MLN-518, lapatinib, gefitinib (iressa), erlotinib (tarceva), perifosine, CYC-202, LY-317615, squalamine, UCN-01, midostaurin, irofulven, staurosporine, alvocidib, genistein, DA-9601, avicine, docetaxel, IM 862, SU 101, and tetrathiomolybdate as well as other agents that induce apoptosis, such as, but not limited to polynucleotides (e.g., ribozymes); polypeptides (e.g., enzymes); drugs; biological mimetics; 25 alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal antibodies conjugated with anticancer drugs, toxins, and/or radionuclides; biological response modifiers (e.g., interferons [e.g., IFN-a, etc.] and interleukins [e.g., IL-2, etc.], etc.); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid, etc.); gene 30 therapy reagents; antisense therapy reagents and nucleotides; tumor vaccines; and inhibitors of angiogenesis, and the like. Numerous other examples of chemotherapeutic compounds and anticancer therapies suitable for co-administration with the disclosed imidazoquinoxaline compounds and analogs thereof will be known and apparent to those skilled in the art.
  • In some embodiments, anticancer agents comprise agents that induce or stimulate apoptosis. Agents that induce apoptosis include, but are not limited to, radiation (e.g., W); kinase inhibitors (e.g., Epidermal Growth Factor Receptor [EGFR] kinase; inhibitor, Vascular Growth Factor Receptor [VGFR] kinase inhibitor, Fibroblast Growth 5 Factor Receptor [FGFR] kinase inhibitor, Platelet-derived Growth Factor Receptor [PGFR] I kinase inhibitor, EGFr and Bcr-Ab1 kinase inhibitors such as Gleevec, Iressa, and Tarceva]); antisense molecules; antibodies [e.g., Herceptin and Rituxan]; anti-estrogens [e.g., raloxifene and tamoxifen]; anti-androgens [e.g., flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids]; cyclooxygenase 2 (COX-2) inhibitors [e.g., Celecoxib, meloxicam, NS-398, and non-steroidal
  • Anti-inflammatory drugs I (NSAIDs)]; and cancer chemotherapeutic drugs [e.g., CPT-11, fludarabine (Fludara), dacarbazine (DTIC), dexamethasone, mitoxantrone, Mylotarg, cisplatinum, 5-FU, Doxrubicin, Taxotere or taxol]; cellular signaling molecules; ceramides and cytokines; and the like may also be administered to subjects in conjunction with the imidazoquinoxalines of Formula (I) or (II).
  • In other embodiments, methods of treating allergies are provided. Such methods include administering an imidazoquinoxaline compound alone or in combination with another agent known to be effective against allergies. In such embodiments, the combination is more effective in treating an allergic condition than the known agcnt(s) is/are without the addition of the imidazoquinoxaline compound. In some such embodiments, the known agent is an antihistamine and/or a leukotriene inhibitor. In other embodiments, the allergic condition is asthma. In other embodiments, the allergic condition is selected from allergic rhinitis, dermatosis, or urticaria. In some such embodiments, the combination is administered to the subject enterally, parenterally, intranasally, subcutaneously, or intraarterially.
  • Compositions contemplated to be within the scope of the present invention may include (an) additional adjuvant(s) and or other immune stimulator compound.
  • Adjuvants
  • Vaccines or immunogenic compositions of the invention may be administered in conjunction with other immunoregulatory agents. In particular, compositions can include an adjuvant. Adjuvants for use with the invention include, but are not limited to, one or more of the following set forth below:
  • Mineral Containing Compositions
  • Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminum salts and calcium salts. The invention includes mineral salts such as hydroxides (e.g. oxyhydroxides), phosphates (e.g. hydroxyphosphates, orthophosphates), sulfates, etc. (e.g. see chapters 8 & 9 of Vaccine Design... (1995) eds. Powell & Newman. ISBN: 030644867X. Plenum.), or mixtures of different mineral compounds (e.g. a mixture of a phosphate and a hydroxide adjuvant, optionally with an excess of the phosphate), with the compounds taking any suitable form (e.g. gel, crystalline, amorphous, etc.), and with adsorption to the salt(s) being preferred. The mineral containing compositions may also be formulated as a particle of metal salt ( WO00/23105 ).
  • Aluminum salts may be included in vaccines of the invention such that the dose of Al3+ is between 0.2 and 1.0 mg per dose.
  • In one embodiment the aluminum based adjuvant for use in the present invention is alum (aluminum potassium sulfate (AlK(SO4)2)), or an alum derivative, such as that formed in-situ by mixing an antigen in phosphate buffer with alum, followed by titration and precipitation with a base such as ammonium hydroxide or sodium hydroxide.
  • Another aluminum-based adjuvant for use in vaccine formulations of the present invention is aluminum hydroxide adjuvant (Al(OH)3) or crystalline aluminum oxyhydroxide (AlOOH), which is an excellent adsorbant, having a surface area of approximately 500m2/g. Alternatively, aluminum phosphate adjuvant (AlPO4) or aluminum hydroxyphosphate, which contains phosphate groups in place of some or all of the hydroxy groups of aluminum hydroxide adjuvant is provided. Preferred aluminum phosphate adjuvants provided herein are amorphous and soluble in acidic, basic and neutral media.
  • In another embodiment the adjuvant of the invention comprises both aluminum phosphate and aluminum hydroxide. In a more particular embodiment thereof, the adjuvant has a greater amount of aluminum phosphate than aluminum hydroxide, such as a ratio of 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1 or greater than 9:1, by weight aluminum phosphate to aluminum hydroxide. More particular still, aluminum salts in the vaccine are present at 0.4 to 1.0 mg per vaccine dose, or 0.4 to 0.8 mg per vaccine dose, or 0.5 to 0.7 mg per vaccine dose, or about 0.6 mg per vaccine dose.
  • Generally, the preferred aluminum-based adjuvant(s), or ratio of multiple aluminum-based adjuvants, such as aluminum phosphate to aluminum hydroxide is selected by optimization of electrostatic attraction between molecules such that the antigen carries an opposite charge as the adjuvant at the desired pH. For example, aluminum phosphate adjuvant (iep = 4) adsorbs lysozyme, but not albumin at pH 7.4. Should albumin be the target, aluminum hydroxide adjuvant would be selected (iep 11.4). Alternatively, pretreatment of aluminum hydroxide with phosphate lowers its isoelectric point, making it a preferred adjuvant for more basic antigens.
  • Oil-Emulsions
  • Oil-emulsion compositions suitable for use as adjuvants in the invention include squalene-water emulsions, such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer). See WO90/14837 . See also, Podda, "The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine", Vaccine (2001) 19: 2673-2680; Frey et al., "Comparison of the safety, tolerability, and immunogenicity of a MF59-adjuvanted influenza vaccine and a nonadjuvanted influenza vaccine in non-elderly adults", Vaccine (2003) 21:4234-4237. MF59 is used as the adjuvant in the FLUAD™ influenza virus trivalent subunit vaccine.
  • Particularly preferred adjuvants for use in the compositions are submicron oil-in-water emulsions. Preferred submicron oil-in-water emulsions for use herein are squalene/water emulsions optionally containing varying amounts of MTP-PE, such as a submicron oil-in-water emulsion containing 4-5% w/v squalene, 0.25-1.0% w/v Tween 80™ (polyoxyelthylenesorbitan monooleate), and/or 0.25-1.0% Span 85™ (sorbitan trioleate), and, optionally, N-acetylmuramyl-L-alanyl-D-isogluatminyl-L-alanine-2-(1'-2'-dipalmitoylsn-glycero-3-huydroxyphosphophoryloxy)-ethylamine (MTP-PE), for example, the submicron oil-in-water emulsion known as "MF59" (International Publication No. WO90/14837 ; US Patent Nos. 6,299,884 and 6,451,325 , and Ott et al., "MF59 -- Design and Evaluation of a Safe and Potent Adjuvant for Human Vaccines" in Vaccine Design: The Subunit and Adjuvant Approach (Powell, M.F. and Newman, M.J. eds.) Plenum Press, New York, 1995, pp. 277-296). MF59 contains 4-5% w/v Squalene (e.g. 4.3%), 0.25-0.5% w/v Tween 80™, and 0.5% w/v Span 85™ and optionally contains various amounts of MTP-PE, formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton, MA). For example, MTP-PE may be present in an amount of about 0-500 µg/dose, more preferably 0-250 µg/dose and most preferably, 0-100 µg/dose. As used herein, the term "MF59-0" refers to the above submicron oil-in-water emulsion lacking MTP-PE, while the term MF59-MTP denotes a formulation that contains MTP-PE. For instance, "MF59-100" contains 100 µg MTP-PE per dose, and so on. MF69, another submicron oil-in-water emulsion for use herein, contains 4.3% w/v squalene, 0.25% w/v Tween 80™, and 0.75% w/v Span 85™ and optionally MTP-PE. Yet another submicron oil-in-water emulsion is MF75, also known as SAF, containing 10% squalene, 0.4% Tween 80™, 5% pluronic-blocked polymer L121, and thr-MDP, also microfluidized into a submicron emulsion. MF75-MTP denotes an MF75 formulation that includes MTP, such as from 100-400 µg MTP-PE per dose.
  • Submicron oil-in-water emulsions, methods of making the same and immunostimulating agents, such as muramyl peptides, for use in the compositions, arc described in detail in International Publication No. WO90/14837 and US Patent Nos. 6,299,884 and 6,45 1,325 .
    Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IFA) may also be used as adjuvants in the invention.
  • Specific oil-in-water emulsion adjuvants useful with the invention include, but are not limited to:
    1. (1) A submicron emulsion of squalene, Tween 80, and Span 85. The composition of the emulsion by volume can be about 5% squalene, about 0.5% polysorbate 80 and about 0.5% Span 85. In weight terms, these ratios become 4.3% squalene, 0.5% polysorbate 80 and 0.48% Span 85. This adjuvant is known as 'MF59' [ WO90/14837 .-Podda & Del Giudice (2003) Expert Rev Vaccines 2:197-203.Podda (2001) Vaccine 19: 2673-2680.], as described in more detail in Chapter 10 of ref. Vaccine Design: The Subunit and Adjuvant Approach (eds. Powell & Newman) Plenum Press 1995 (ISBN 0-306-44867-X ). and chapter 12 of ref. Vaccine Adjuvants: Preparation Methods and Research Protocols (Volume 42 of Methods in Molecular Medicine series). ISBN: 1-59259-083-7. Ed. O'Hagan. The MF59 emulsion advantageously includes citrate ions e.g. 10mM sodium citrate buffer.
    2. (2) An emulsion of squalene, a tocopherol, and Tween 80. The emulsion may include phosphate buffered saline. It may also include Span 85 (e.g. at 1%) and/or lecithin. These emulsions may have from 2 to 10% squalene, from 2 to 10% tocopherol and from 0.3 to 3% Tween 80, and the weight ratio of squalene:tocopherol is preferably ≤1 as this provides a more stable emulsion. One such emulsion can be made by dissolving Tween 80 in PBS to give a 2% solution, then mixing 90ml of this solution with a mixture of (5g of DL-α-tocopherol and 5ml squalene), then microfluidising the mixture. The resulting emulsion may have submicron oil droplets e.g. with an average diameter of between 100 and 250nm, preferably about 180nm.
    3. (3) An emulsion of squalene, a tocopherol, and a Triton detergent (e.g. Triton X-100).
    4. (4) An emulsion of squalane, polysorbate 80 and poloxamer 401 ("Pluronic™ L121"). The emulsion can be formulated in phosphate buffered saline, pH 7.4. This emulsion is a useful delivery vehicle for muramyl dipeptides, and has been used with threonyl-MDP in the "SAF-1" adjuvant [Allison & Byars (1992) Res Immunol 143:519-25] (0.05-1% Thr-MDP, 5% squalane, 2.5% Pluronic L121 and 0.2% polysorbate 80). It can also be used without the Thr-MDP, as in the "AF" adjuvant [Hariharan et al. (1995) Cancer Res 55:3486-9] (5% squalane, 1.25% Pluronic L121 and 0.2% polysorbate 80). Microfluidisation is preferred.
  • The emulsions are preferably mixed with additional agents (such as an antigen) extemporaneously, at the time of delivery. Thus the adjuvant and antigen are typically kept separately in a packaged or distributed vaccine, ready for final formulation at the time of use. The antigen will generally be in an aqueous form, such that the vaccine is finally prepared by mixing two liquids. The volume ratio of the two liquids for mixing can vary (e.g. between 5:1 and 1:5) but is generally about 1:1.
  • Where a composition includes a tocopherol, any of the α, β, γ, δ, ε or ξ tocopherols can be used, but α-tocopherols are preferred. The tocopherol can take several forms e.g. different salts and/or isomers. Salts include organic salts, such as succinate, acetate, nicotinate, etc. D-α-tocopherol and DL-α-tocopherol can both be used. Tocopherols are advantageously included in vaccines for use in elderly patients (e.g. aged 60 years or older) because vitamin E has been reported to have a positive effect on the immune response in this patient group [Han et al. (2005) Impact of Vitamin E on Immune Function and Infectious Diseases in the Aged at Nutrition, Immune functions and Health EuroConference, Paris, 9-10 June 2005]. They also have antioxidant properties that may help to stabilize the emulsions [ US- 6630161 ]. A preferred α-tocopherol is DL-α-tocopherol, and the preferred salt of this tocopherol is the succinate. The succinate salt has been found to cooperate with TNF-related ligands in vivo. Moreover, α-tocopherol succinate is known to be compatible with influenza vaccines and to be a useful preservative as an alternative to mercurial compounds
  • Saponin Formulations
  • Saponin formulations, may also be used as adjuvants in the invention. Saponins are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponins isolated from the bark of the Quillaia saponaria Molina tree have been widely studied as adjuvants. Saponins can also be commercially obtained from Smilax ornata (sarsaprilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root). Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
  • Saponin compositions have been purified using High Performance Thin Layer Chromatography (HP-TLC) and Reversed Phase High Performance Liquid Chromatography (RP-HPLC). Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C. Preferably, the saponin is QS21. A method of production of QS21 is disclosed in US Patent No. 5,057,540 . Saponin formulations may also comprise a sterol, such as cholesterol (see WO96/33739 ).
  • Combinations of saponins and cholcstcrols can be used to form unique particles called Immunostimulating Complexes (ISCOMs). ISCOMs typically also include a phospholipid such as phosphatidylethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs. Preferably, the ISCOM includes one or more of Quil A, QHA and QHC. ISCOMs are further described in EP0109942 , WO96/11711 and WO96/33739 . Optionally, the ISCOMS may be devoid of (an) additional detergent(s). See WO00/07621 .
  • A review of the development of saponin based adjuvants can be found in Barr, et al., "ISCOMs and other saponin based adjuvants", Advanced Drug Delivery Reviews (1998) 32:247-271. See also Sjolander, et al., "Uptake and adjuvant activity of orally delivered saponin and ISCOM vaccines", Advanced Drug Delivery Reviews (1998) 32:321-338.
  • Virosomes and Virus Like Particles (VLPs)
  • Virosomes and Virus Like Particles (VLPs) can also be used as adjuvants in the invention. These structures generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally non-pathogenic, nonreplicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses. These viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA), Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and-Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA-phages, Qβ-phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein p1). VLPs are discussed further in WO03/024480 , WO03/024481 , and Niikura et al., "Chimeric Recombinant Hepatitis E Virus-Like Particles as an Oral Vaccine Vehicle Presenting Foreign Epitopes", Virology (2002) 293:273-280; Lenz et al., "Papillomarivurs-Like Particles Induce Acute Activation of Dendritic Cells", Journal of Immunology (2001) 5246-5355 ; Pinto, et al., "Cellular Immune Responses to Human Papillomavirus (HPV)-16 L1 Healthy Volunteers Immunized with Recombinant HPV-16 L1 Virus-Like Particles", Journal of Infectious Diseases (2003) 188:327-338; and Gerber et al., "Human Papillomavirus Virus-Like Particles Are Efficient Oral Immunogens when Coadministered with Escherichia coli Heat-Labile Entertoxin Mutant R192G or CpG", Journal of Virology (2001) 75(10):4752-4760. Virosomes are discussed further in, for example, Gluck et al., "New Technology Platforms in the Development of Vaccines for the Future", Vaccine (2002) 20:B10 -B16. Immunopotentiating reconstituted influenza virosomes (IRIV) are used as the subunit antigen delivery system in the intranasal trivalent INFLEXAL™ product {Mischler & Metcalfe (2002) Vaccine 20 Suppl 5:B17-23} and the INFLUVAC PLUS™ product.
  • Bacterial or Microbial Derivatives
  • Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as:
  • (1) Non-toxic derivatives of enterobacterial lipopolysaccharide (LPS)
  • Such derivatives include Monophosphoryl lipid A (MPL) and 3-O-deacylated MPL (3dMPL). 3dMPL is a mixture of 3 De-O-acylated monophosphoryl lipid A with 4, 5 or 6 acylated chains. A preferred "small particle" form of 3 De-O-acylated monophosphoryl lipid A is disclosed in EP 0 689 454 . Such "small particles" of 3dMPL are small enough to be sterile filtered through a 0.22 micron membrane (see EP 0 689 454 ). Other non-toxic LPS derivatives include monophosphoryl lipid A mimics, such as aminoalkyl glucosaminide phosphate derivatives e.g. RC-529. See Johnson et al. (1999) Bioorg Med Chem Lett 9:2273-2278.
  • 3dMPL has been prepared from a heptoseless mutant of Salmonella minnesota. It activates cells of the monocyte/macrophage lineage and stimulates release of several cytokines, including IL-1, IL-12, TNF-α and GM-CSF (see also ref. Thompson et al. (2005) J Leukoc Biol 78: 'The low-toxicity versions of LPS, MPL® adjuvant and RC529, are efficient adjuvants for CD4+ T cells'.). Preparation of 3dMPL was originally described in reference UK patent application GB-A-2220211 .
  • 3dMPL can take the form of a mixture of related molecules, varying by their acylation (e.g. having 3, 4, 5 or 6 acyl chains, which may be of different lengths). The two glucosamine (also known as 2-deoxy-2-amino-glucose) monosaccharides are N-acylated at their 2-position carbons (i.e. at positions 2 and 2'), and there is also O-acylation at the 3' position. The group attached to carbon 2 has formula -NH-CO-CH2-CR1R1'. The group attached to carbon 2' has formula -NH-CO-CH2-CR2R2'. The group attached to carbon 3' has formula -O-CO-CH2-CR3R3'. A representative structure is:
    Figure imgb0016
  • Groups R1, R2 and R3 are each independently -(CH2)n-CH3. The value of n is preferably between 8 and 16, more preferably between 9 and 12, and is most preferably 10. Groups R1', R2' and R3' can each independently be: (a) -H; (b) hydroxy; or (c) - O-CO-R4,where R4 is either -H or -(CH2)m-CH3, wherein the value of m is preferably between 8 and 16, and is more preferably 10, 12 or 14. At the 2 position, m is preferably 14. At the 2' position, m is preferably 10. At the 3' position, m is preferably 12. Groups R1', R2' and R3' are thus preferably -O-acyl groups from dodecanoic acid, tetradecanoic acid or hexadecanoic acid.
  • When all of R1', R2' and R3' are -H then the 3dMPL has only 3 acyl chains (one on each of positions 2, 2' and 3'). When only two of R1', R2' and R3' are -H then the 3dMPL can have 4 acyl chains. When only one of R1', R2' and R3' is -H then the 3dMPL can have 5 acyl chains. When none of R1', R2' and R3' is -H then the 3dMPL can have 6 acyl chains. The 3dMPL adjuvant used according to the invention can be a mixture of these forms, with from 3 to 6 acyl chains, but it is preferred to include 3dMPL with 6 acyl chains in the mixture, and in particular to ensure that the hexaacyl chain form makes up at least 10% by weight of the total 3dMPL e.g. ≥20%, ≥30%, ≥40%, ≥50% or more. 3dMPL with 6 acyl chains has been found to be the most adjuvant-active form.
  • Thus the most preferred form of 3dMPL for inclusion in compositions of the invention is:
    Figure imgb0017
  • Where 3dMPL is used in the form of a mixture then references to amounts or concentrations of 3dMPL in compositions of the invention refer to the combined 3dMPL species in the mixture.
  • In aqueous conditions, 3dMPL can form micellar aggregates or particles with different sizes e.g. with a diameter <150 nm or >500 nm. Either or both of these can be used with the invention, and the better particles can be selected by routine assay. Smaller particles (e.g. small enough to give a clear aqueous suspension of 3dMPL) are preferred for use according to the invention because of their superior activity [ WO 94/21292 ]. Preferred particles have a mean diameter less than 220 nm, more preferably less than 200 nm or less than 150 nm or less than 120 nm, and can even have a mean diameter less than 100 nm. In most cases, however, the mean diameter will not be lower than 50 nm. These particles are small enough to be suitable for filter sterilization. Particle diameter can be assessed by the routine technique of dynamic light scattering, which reveals a mean particle diameter. Where a particle is said to have a diameter of x nm, there will generally be a distribution of particles about this mean, but at least 50% by number (e.g. ≥60%, ≥70%, ≥80%, ≥90%, or more) of the particles will have a diameter within the range x±25%.
  • 3dMPL can advantageously be used in combination with an oil-in-water emulsion. Substantially all of the 3dMPL may be located in the aqueous phase of the emulsion. The 3dMPL can be used on its own, or in combination with one or more further compounds. For example, it is known to use 3dMPL in combination with the QS21 saponin [ WO94/00153 .] (including in an oil-in-water emulsion [ WO95/17210 ]), with an immunostimulatory oligonucleotide, with both QS21 and an immunostimulatory oligonucleotide, with aluminum phosphate [ WO96/26741 ], with aluminum hydroxide [ WO93/19780 ], or with both aluminum phosphate and aluminum hydroxide.
  • Lipid A Derivatives
  • Lipid A derivatives include derivatives of lipid A from Escherichia coli such as OM-174. OM-174 is described for example in Meraldi et al., "OM-174, a New Adjuvant with a Potential for Human Use, Induces a Protective Response with Administered with the Synthetic C-Terminal Fragment 242-310 from the circumsporozoite protein of Plasmodium berghei", Vaccine (2003) 21:2485-2491; and Pajak, et al., "The Adjuvant OM-174 induces both the migration and maturation of murine dendritic cells in vivo", Vaccine (2003) 21:836-842 .
  • Immunostimulatory oligonucleotides
  • Immunostimulatory oligonucleotides suitable for use as adjuvants in the invention include nucleotide sequences containing a CpG motif (a sequence containing an unmethylated cytosine followed by guanosine and linked by a phosphate bond). Bacterial double stranded RNA or oligonucleotides containing palindromic or poly(dG) sequences have also been shown to be immunostimulatory.
  • The CpG's can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or single-stranded. Optionally, the guanosine may be replaced with an analog such as 2'-deoxy-7-deazaguanosine. See Kandimalla, et al., "Divergent synthetic nucleotide motif recognition pattern: design and development of potent immunomodulatory oligodeoxyribonucleotide agents with distinct cytokine induction profiles", Nucleic Acids Research (2003) 31(9): 2393-2400; WO02/26757 and WO99/62923 for examples of possible analog substitutions. The adjuvant effect of CpG oligonucleotides is further discussed in Krieg, "CpG motifs: the active ingredient in bacterial extracts?", Nature Medicine (2003) 9(7): 831-835; McCluskie, et al., "Parenteral and mucosal prime-boost immunization strategies in mice with hepatitis B surface antigen and CpG DNA", FEMS Immunology and Medical Microbiology (2002) 32:179-185; WO98/40100 ; US Patent No. 6,207,646 ; US Patent No. 6,239,116 and US Patent No. 6,429,199 .
  • The CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT. See Kandimalla, et al., "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic CpG DNAs", Biochemical Society Transactions (2003) 31 (part 3): 654-658. The CpG sequence may be specific for inducing a Th1 immune response, such as a CpG-A ODN, or it may be more specific for inducing a B cell response, such a CpG-B ODN. CpG-A and CpG-B ODNs are discussed in Blackwell, et al., "CpG-A-Induced Monocyte IFN-gamma-Inducible Protein-10 Production is Regulated by Plasmacytoid Dendritic Cell Derived IFN-alpha", J. Immunol. (2003) 170(8):4061-4068; Krieg, "From A to Z on CpG", TRENDS in Immunology (2002) 23(2): 64-65 and WO01/95935 . Preferably, the CpG is a CpG-A ODN.
  • Examples of CpG nucleotides include the following sequences, which may contain phosphorothioate modified internucleotide linkages:
    Figure imgb0018
    See WO 05/25614 .
  • Preferably, the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition. Optionally, two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers". See, for example, Kandimalla, et al., "Secondary structures in CpG oligonucleotides affect immunostimulatory activity", BBRC (2003) 306:948-953; Kandimalla, et al., "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic GpG DNAs", Biochemical Society Transactions (2003) 31(part 3):664-658 ; Bhagat et al., "CpG penta- and hexadeoxyribonucleotides as potent immunomodulatory agents" BBRC (2003) 300:853-861 and WO03/035836 .
  • ADP-ribosylating toxins and detoxified derivatives thereof.
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention. Preferably, the protein is derived from E. coli (i.e., E. coli heat labile enterotoxin "LT), cholera ("CT"), or pertussis ("PT"). The use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in WO95/17211 and as parenteral adjuvants in WO98/42375 . Preferably, the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LTR192G. The use of ADP-ribosylating toxins and detoxified derivatives thereof, particularly LT-K63 and LT-R72, as adjuvants can be found in the following references: Beignon, et al., "The LTR72 Mutant of Heat-Labile Enterotoxin of Escherichia coli Enhances the Ability of Peptide Antigens to Elicit CD4+ T Cells and Secrete Gamma Interferon after Coapplication onto Bare Skin", Infection and Immunity (2002) 70(6):3012-3019; Pizza, et al., "Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants", Vaccine (2001) 19:2534-2541; Pizza, et al., "LTK63 and LTR72, two mucosal adjuvants ready for clinical trials" Int. J. Med. Microbiol (2000) 290(4-5):455-461; Scharton-Kersten et al., "Transcutaneous Immunization with Bacterial ADP-Ribosylating Exotoxins, Subunits and Unrelated Adjuvants", Infection and Immunity (2000) 68(9):5306-5313 ; Ryan et al., "Mutants of Escherichia coli Heat-Labile Toxin Act as Effective Mucosal Adjuvants for Nasal Delivery of an Acellular Pertussis Vaccine: Differential Effects of the Nontoxic AB Complex and Enzyme Activity on Th1 and Th2 Cells" Infection and Immunity (1999) 67(12):6270-6280; Partidos et al., "Heat-labile enterotoxin of Escherichia coli and its site-directed mutant LTK63 enhance the proliferative and cytotoxic T-cell responses to intranasally co-immunized synthetic peptides", Immunol. Lett. (1999) 67(3):209-216; Peppoloni et al., "Mutants of the Escherichia coli heat-labile enterotoxin as safe and strong adjuvants for intranasal delivery of vaccines", Vaccines (2003) 2(2):285-293; and Pine et al., (2002) "Intranasal immunization with influenza vaccine and a detoxified mutant of heat labile enterotoxin from Escherichia coli (LTK63)" J. Control Release (2002) 85(1-3):263-270. Numerical reference for amino acid substitutions is preferably based on the alignments of the A and B subunits of ADP-ribosylating toxins set forth in Domenighini et al., Mol. Microbiol (1995) 15(6):1165-1167. Bioadhesives and Mucoadhesives
  • Bioadhesives and mucoadhesives may also be used as adjuvants in the invention. Suitable bioadhesives include esterified hyaluronic acid microspheres (Singh et al. (2001) J. Cont. Rele. 70:267-276) or mucoadhesives such as cross-linked derivatives of polyacrylic acid, polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides and carboxymethylcellulose. Chitosan and derivatives thereof may also be used as adjuvants in the invention. E.g. WO99/27960 .
  • Microparticles
  • Microparticles may also be used as adjuvants in the invention. Microparticles (i.e. a particle of ~100nm to ~150µm in diameter, more preferably ~200nm to ~30µm in diameter, and most preferably ~500nm to ~10µm in diameter) formed from materials that are biodegradable and non-toxic (e.g. a poly(α-hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.), with poly(lactide-co-glycolide) are preferred, optionally treated to have a negatively-charged surface (e.g. with SDS) or a positively-charged surface (e.g. with a cationic detergent, such as CTAB).
  • Liposomes
  • Examples of liposome formulations suitable for use as adjuvants are described in US Patent No. 6,090,406 , US Patent No. 5,916,588 , and EP 0 626 169 .
  • Polyoxyethylene ether and Polyoxyethylene Ester Formulations
  • Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters. WO99/52549 . Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol ( WO01/21207 ) as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol ( WO01/21152 ).
  • Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether (laureth 9), polyoxyethylene-9-steoryl ether, polyoxytheylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
  • Polyphosphazene (PCPP)
  • PCPP formulations are described, for example, in Andrianov et al., "Preparation of hydrogel microspheres by coacervation of aqueous polyphophazene solutions", Biomaterials (1998) 19(1-3):109-115 and Payne et al., "Protein Release from Polyphosphazene Matrices", Adv. Drug. Delivery Review (1998) 31(3):185-196.
  • Muramyl peptides
  • Examples of muramyl peptides suitable for use as adjuvants in the invention include N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-1-alanyl-d-isoglutamine (nor-MDP), and N-acetylmuramyl-1-alanyl-d-isoglutaminyl-1-alanine-2-(1'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-ethylamine MTP-PE).
  • Small Molecule Immunopontentiators (SMIPs) Imidazoquinoline Compounds.
  • Examples of imidazoquinoline compounds suitable for use adjuvants in the invention include Imiquimod and its analogues, described further in Stanley, "Imiquimod and the imidazoquinolines: mechanism of action and therapeutic potential" Clin Exp Dermatol (2002) 27(7):571-577; Jones, "Resiquimod 3M", Curr Opin Investig Drugs (2003) 4(2):214-218; Wu et al. (2004) Antiviral Res. 64(2):79-83 Vasilakos et al. (2000) Cell Immunol. 204(1):64-74 US patents 4689338 , 4929624 , 5238944 , 5266575 , 5268376 , 5346905 , 5352784 , 5389640 , 5395937 , 5482936 , 5494916 , 5525612 , 6083505 , 6440992 , 6627640 , 6656938 , 6660735 , 6660747 , 6664260 , 6664264 , 6664265 , 6667312 , 6670372 , 6677347 , 6677348 , 6677349 , 6683088 , 6703402 , 6743920 , 6800624 , 6809203 , 6888000 and 6924293 .
  • Preferred SMIPs include:
    • N2-methyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2,N2-dimethyl-1-(2-methylpropyl)-1H-imidazo [4,5-c]quinoline-2,4-diamine;
    • N2-ethyl-N2-methyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-methyl-1-(2-methylpropyl)-N2-propyl-1H-imidazo [4,5-c] quinoline-2,4-diamine;
    • 1-(2-methylpropyl)-N2-propyl-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-butyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-butyl-N2-methyl-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-methyl-1-(2-methylpropyl)-N2-pentyl-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-methyl-1-(2-methylpropyl)-N2-prop-2-enyl-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • 1-(2-methylpropyl)-2-[(phenylmethyl)thio]-1H-imidazo[4,5-c]quinolin-4-amine;
    • 1-(2-methylpropyl)-2-(propylthio)-1H-imidazo[4,5-c]quinolin-4-amine ;
    • 2-[[4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl](methyl)amino] ethanol;
    • 2-[[4-amino-1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-2-yl](methyl)amino]ethyl acetate;
    • 4-amino-1-(2-methylpropyl)-1,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one;
    • N2-butyl-1-(2-methylpropyl)-N4,N4-bis(phenylmethyl)-1H-imidazo[4,5-c] quinoline-2,4-diamine;
    • N2-butyl-N2-methyl-1-(2-methylpropyl)-N4,N4-bis(phenylmethyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2-methyl-1-(2-methylpropyl)-N4,N4-bis(phenylmethyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • N2,N2-dimethyl-1-(2-methylpropyl)-N4,N4-bis(phenylmethyl)-1H-imidazo[4,5-c]quinoline-2,4-diamine;
    • 1-{4-amino-2-[methyl(propyl)amino]-1H-imidazo[4,5-c]quinolin-1-yl}-2-methylpropan-2-ol;
    • 1-[4-ammo-2-(propylammo)-1H-imidazo[4,5-c]quinolin-1-yl]-2-methylpropan-2-ol;
    • N4,N4-dibenzyl-1-(2-methoxy-2-methylpropyl)-N2-propyl-1H-imidazo[4,5-c]quinoline-2,4-diamine.
    Nucleoside Analogs.
  • A nucleoside analog, such as: (a) Isatorabine (ANA-245; 7-thia-8-oxoguanosine):
    Figure imgb0019
    and prodrugs thereof; (b)ANA975; (c) ANA-025-1; (d) ANA380; (e) the compounds disclosed in references US 6,924,271 to US2005/0070556 US 5,658,731 ; (f) a compound having the Formula:
    Figure imgb0020
    wherein:
    • R1 and R2 are each independently H, halo, -NRaRb, hydroxy, C1-6 alkoxy, substituted C1-6 alkoxy, heterocyclyl, substituted heterocyclyl, C6-10 aryl, substituted C6-10 aryl, C1-6 alkyl, or substituted C1-6 alkyl;
    • R3 is absent, H, C1-6 alkyl, substituted C1-6 alkyl, C6-10 aryl, substituted C6-10 aryl, heterocyclyl, or substituted heterocyclyl;
    • R4 and R5 are each independently H, halo, heterocyclyl, substituted heterocyclyl, -C(O)-Rd, C1-6 alkyl, substituted C1-6 alkyl, or bound together to form a 5 membered ring as in R4-5:
      Figure imgb0021
      the binding being achieved at the bonds indicated by a
      Figure imgb0022
    • X1 and X2 are each independently N, C, O, or S;
    • R8 is H, halo, hydroxy, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, hydroxy, - NRaRb, -(CH2)n-O-Rc, -O-(C1-6 alkyl), -S(O)pRe, or -C(O)-Rd;
    • R9 is H, C1-6 alkyl, substituted C1-6 alkyl, heterocyclyl, substituted heterocyclyl or R9a, wherein R9a is:
      Figure imgb0023
      the binding being achieved at the bond indicated by a
      Figure imgb0024
    • R10 and R11 are each independently H, halo, C1-6 alkoxy, substituted C1-6 alkoxy, -NRaRb, or hydroxy;
    • each Ra and Rb is independently H, C1-6 alkyl, substituted C1-6 alkyl, - C(O)Rd, C6-10 aryl;
    • each Rc is independently H, phosphate, diphosphate, triphosphate, C1-6 alkyl, or substituted C1-6 alkyl;
    • each Rd is independently H, halo, C1-6 alkyl, substituted C1-6 alkyl, C1-6 alkoxy, substituted C1-6 alkoxy, -NH2, -NH(C1-6 alkyl), -NH(substituted C1-6 alkyl), -N(C1-6 alkyl)2, -N(substituted C1-6 alkyl)2, C6-10 aryl, or heterocyclyl;
    • each Re is independently H, C1-6 alkyl, substituted C1-6 alkyl, C6-10 aryl, substituted C6-10 aryl, heterocyclyl, or substituted heterocyclyl;
    • each Rf is independently H, C1-6 alkyl, substituted C1-6 alkyl, -C(O)Rd, phosphate, diphosphate, or triphosphate;
    • each n is independently 0, 1, 2, or 3;
    • each p is independently 0, 1, or 2; or
    or (g) a pharmaceutically acceptable salt of any of (a) to (f), a tautomer of any of (a) to (f), or a pharmaceutically acceptable salt of the tautomer;
    Loxoribine (7-allyl-8-oxoguanosine) [ US patent 5,011,828 ]. Thiosemicarbazone Compounds.
  • Examples of thiosemicarbazone compounds, as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in WO04/60308 . The thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclcar cells for the production of cytokines, such as TNF-α.
  • Tryptanthrin Compounds.
  • Examples of tryptanthrin compounds, as well as methods of formulating, manufacturing, and screening for compounds all suitable for use as adjuvants in the invention include those described in WO04/64759 . The tryptanthrin compounds are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF- α.
  • Additional SMIPs
    1. (i) Compounds disclosed in reference W02004/87153 , including: Acylpiperazine compounds, Indoledione compounds, Tetrahydraisoquinoline (THIQ) compounds, Benzocyclodione compounds, Aminoazavinyl compounds, Aminobenzimidazole quinolinone (ABIQ) compounds [ US 6,605,617 , WO02/18383 ], Hydrapthalamide compounds, Benzophenone compounds, Isoxazole compounds, Sterol compounds, Quinazilinone compounds, Pyrrole compounds [ WO2004/018455 ], Anthraquinone compounds, Quinoxaline compounds, Triazine compounds, Pyrazalopyrimidine compounds, and Benzazole compounds [ WO03/082272 ].
    2. (ii) Methyl inosine 5'-monophosphate ("MIMP") [Signorelli & Hadden (2003) Int Immunopharmacol 3(8):1177-86.].
    3. (iii) A polyhydroxlated pyrrolizidine compound [ WO2004/064715 ], such as one having Formula:
      Figure imgb0025
      where R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof. Examples include, but are not limited to: casuarine, casuarine-6-α-D-glucopyranose, 3-epi-casuarine, 7-epi-casuarine, 3,7-diepi-casuarine, etc.
    4. (iv) A gamma inulin [Cooper (1995) Pharm Biotechnol 6:559-80] or derivative thereof, such as algammulin.
    Human Immunomodulators
  • Human immunomodulators suitable for use as adjuvants in the invention include cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g. interferon-γ), macrophage colony stimulating factor, and tumor necrosis factor.
  • Aluminum salts and MF59 are preferred adjuvants for use with injectable i vaccines. Bacterial toxins and bioadhesives are preferred adjuvants for use with mucosally-delivered vaccines, such as nasal vaccines.
  • TLR Modulators/Agonists
  • By "TLR agonist" it is meant a component which is capable of causing a signalling response through a TLR signalling pathway, either as a direct ligand or indirectly through generation of endogenous or exogenous ligand (Sabroe et al, J1 2003 p1630-5). TLR agonists of the present invention, include agonists of the following:
    1. (1) TLR1: Tri- acylated lipopeptides (LPs); phenol-soluble modulin; Mycobacterium tuberculosis LP; S-(2,3-bis(palmitoyloxy)-(2-RS)-propyl)-N-palmitoyl-(R)- Cys-(S)-Ser-(S) Lys(4)-OH, trihydrochloride (Pam3Cys) LP which mimics the acetylated amino terminus of a bacterial lipoprotein and OspA LP from Borrelia burgdorfei);
    2. (2) TLR2: one or more of a bacterial lipopeptide from M tuberculosis, B burgdorferi. T pallidum; peptidoglycans from species including Staphylococcus aureus; lipoteichoic acids, mannuronic acids, Neisseria porins, bacterial fimbriae, Yersina virulence factors, CMV virions, measles haemagglutinin, and zymosan from yeast;
    3. (3) TLR3: double stranded RNA, or polyinosinic- polycytidylic acid (Poly IC), a molecular nucleic acid pattern associated with viral infection;
    4. (4) TLR4: one or more of a lipopolysaccharide (LPS) from gram-negative bacteria, or fragments thereof; heat shock protein (HSP) 10, 60, 65, 70, 75 or 90; surfactant Protein A, hyaluronan oligosaccharides, heparan sulphate fragments, fibronectin fragments, fibrinogen peptides and b-defensin-2. In one embodiment the TLR agonist is HSP 60, 70 or 90. In an alternative embodiment, the TLR agonist capable of causing a signalling response through TLR-4 is a non-toxic derivative of LPS. Monophosphoryl lipid A (MPL) and 3D-MPL as described above, is one such non-toxic derivative. Futher adjuvants and TLR4 modulators include lipids linked to a phosphate-containing acyclic backbone, such as the TLR4 antagonist E5564 [Wong et al. (2003) J Clin Pharmacol 43(7):735-42, US2005/0215517 ]:
      Figure imgb0026
    5. (5) TLR5: including bacterial flagellin;
    6. (6) TLR6: including mycobacterial lipoprotein, di-acylated LP, and phenol-soluble modulin. Further TLR6 agonists are I described in W02003043572 ;
    7. (7) TLR7: including loxoribine, a guanosine analogue at positions N7 and C8, isatoribine, ANA-971, ANA-975, or an imidazoquinoline compound, or derivative thereof. In one embodiment, the TLR agonist is imiquimod or resiquimod. Further TLR7 agonists are described in W002085905 ;
    8. (8) TLR8: an imidazoquinoline molecule, for example resiquimod (R848); resiquimod is also capable of recognition by TLR-7. Other TLR-8 agonists which may be used include those described in W02004071459 ; and/or
    9. (9) TLR9: In one embodiment,, I the TLR agonist capable of causing a signalling response through TLR-9 is HSP90 or a DNA containing unmethylated CpG nucleotide, in particular sequence contexts described above with CpG motifs.
  • Preferred TLR modulators are agonists of TLR7 (e.g. imidazoquinolines) and/or TLR9 (e.g. CpG oligonucleotides).
  • Phospho-containing lipids
  • Compounds disclosed in reference PCT/US2005/022769 .
  • Phosphatidylcholine derivatives and phosphorylcholine containing molecules.
  • A compound of Formula (I), (II) or (III), or a salt thereof:
    Figure imgb0027
    Figure imgb0028
    as defined in reference WO03/011223 , such as 'ER 803058', 'ER 803732', 'ER 804053', ER 804058', 'ER 804059', 'ER 804442', 'ER 804680', 'ER 804764', ER 803022 or `ER 804057' e.g.:
    Figure imgb0029
    Figure imgb0030
  • An aminoalkyl glucosaminide phosphate derivative, such as RC-529 [Johnson et al. (1999) Bioorg Med Chem Lett 9:2273-2278, Evans et al. (2003) Expert Rev Vaccines 2:219-229].
  • The invention may also comprise combinations of aspects of one or more of the adjuvants identified above. For example, the following adjuvant compositions may be used in the invention:
    • (1) a saponin and an oil-in-water emulsion ( WO99/11241 );
    • (2) a saponin (e.g.., QS21) + a non-toxic LPS derivative (e.g. 3dMPL) (see WO94/00153 );
    • (3) a saponin (e.g.., QS21) + a non-toxic LPS derivative (e.g. 3dMPL) + a cholesterol;
    • (4) a saponin (e.g. QS21) + 3dMPL + IL-12 (optionally + a sterol) ( WO98/57659 );
    • (5) combinations of 3dMPL with, for example, QS21 and/or oil-in-water emulsions (See European patent applications 0835318 , 0735898 and 0761231 );
    • (6) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-block polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion.
    • (7) Ribi adjuvant system (RAS), (Ribi Immunochem) containing 2% Squalene, 0.2% Twccn 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL + CWS (Detox™); and
    • (8) one or more mineral salts (such as an aluminum salt) + a non-toxic derivative of LPS (such as 3dPML).
    • (9) (9) one or more mineral salts (such as an aluminum salt) + an immunostimulatory oligonucleotide (such as a nucleotide sequence including a CpG motif).
  • The adjuvants described herein can be added to the composition at various stages during their production. For example, the adjuvant may be within or surround an antigen composition, and this mixture can then be/added to an oil-in-water emulsion. As an alternative, the antigen and/adjuvant may be within an oil-in-water emulsion, in which case the agent can either be added to the emulsion components before emulsification, or it can be added to the emulsion after emulsification. Similarly, the agent may be coaccrvatcd within the emulsion droplets. The location and distribution of the adjuvant within the final composition will depend on its hydrophilic/lipophilic properties e.g. the agent can be located in the aqueous phase, in the oil phase, and/or at the oil-water interface.
  • Further, the adjuvant described herein can be conjugated to a separate agent, such as an antigen (e.g. CRM197) or directly to any amenable composition of the present invention. A general review of conjugation techniques for small molecules is provided in Thompson et al. (2003) Methods in Molecular Medicine 94:255-266. Preferred conjugation methods involve directly coupling through reductive amination or via a linker, such as adipic acid or squarate. As an alternative, the adjuvants may be non-covalently associated with additional agents, such as by way of hydrophobic or ionic interactions.
  • The invention is also directed to methods of administering the immunogenic compositions of the invention, wherein the immunogenic composition can include in one embodiment one or more adjuvnats and antigens as described herein in combination with a compound of Formula (I) or (II). In some embodiments, the immunogenic composition is administered to the subject in an amount effective to stimulate an immune response. The amount that constitutes an effective amount depends, inter alia, on the particular immunogenic composition used, the particular adjuvant compound being administered and the amount thereof, the immune response that is to be enhanced (humoral or cell mediated), the state of the immune system (e.g., suppressed, compromised, stimulated), and the desired therapeutic result. Accordingly it is not practical to set forth generally the amount that constitutes an effective amount of the immunogenic composition. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors.
  • The compositions of the invention can be administered to various animals subjects including mammals such as human and non-human subjects, including, for example, pocket pets, fowl, and the like according to conventional methods well-known to those skilled in the art.
  • The immunogenic compositions of the present invention can be used in the manufacture of a vaccine. Suitable vaccines include, but are not limited to, any material that raises either or both humoral or cell mediated immune response. Suitable vaccines can include live viral and bacterial antigens and inactivated viral, tumor-derived, protozoal, organism-derived, fungal, and bacterial antigens, toxoids, toxins, polysaccharides, proteins, glycoproteins, peptides, and the like, numerous examples of which are described below.
  • Antigens:
  • Compositions of the invention may be administered in conjunction with one or more antigens for use in therapeutic, prophylactic, or diagnostic methods of the present invention. Preferred antigens include those listed below. Additionally, the compositions of the present invention may be used to treat or prevent infections caused by any of the below-listed pathogens. In addition to combination with the antigens described below, the compositions of the invention may also be combined with an adjuvant as described herein.
  • Antigens for use with the invention include, but are not limited to, one or more of the following antigens set forth below, or antigens derived from one or more of the pathogens set forth below:
  • A. Bacterial Antigens
  • Bacterial antigens suitable for use in the invention include proteins, polysaccharides, lipopolysaccharides, and outer membrane vesicles which may be isolated, purified or derived from a bacteria. In addition, bacterial antigens may include bacterial lysates and inactivated bacteria formulations. Bacteria antigens may be produced by recombinant expression. Bacterial antigens preferably include epitopes which are exposed on the surface of the bacteria during at least one stage of its life cycle. Bacterial antigens are preferably conserved across multiple serotypes. Bacterial antigens include antigens derived from one or more of the bacteria set forth below as well as the specific antigens examples identified below.
  • Neisseria meningitides: Meningitides antigens may include proteins (such as those identified in References 1 - 7), saccharides (including a polysaccharide, oligosaccharide or lipopolysaccharide), or outer-membrane vesicles (References 8, 9, 10, 11) purified or derived from N. meningitides serogroup such as A, C, W135, Y, and/or B. Meningitides protein antigens may be selected from adhesions, autotransporters, toxins, Fe acquisition proteins, and membrane associated proteins (preferably integral outer membrane protein).
  • Streptococcus pneumoniae: Streptococcus pneumoniae antigens may include a saccharide (including a polysaccharide or an oligosaccharide) and/or protein from Streptococcus pneumoniae. Saccharide antigens may be selected from serotypes 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F, and 33F. Protein antigens may be selected from a protein identified in WO 98/18931 , WO 98/18930 , US Patent No. 6,699,703 , US Patent No. 6,800,744 , WO 97/43303 , and WO 97/37026 . Streptococcus pneumoniae proteins may be selected from the Poly Histidine Triad family (PhtX), the Choline Binding Protein family (CbpX), CbpX truncates, LytX family, LytX truncates, CbpX truncate-LytX truncate chimeric proteins, pneumolysin (Ply), PspA, PsaA, Sp128, Sp101, Sp130, Sp125 or Sp133.
  • Streptococcus pyogenes (Group A Streptococcus): Group A Streptococcus antigens may include a protein identified in WO 02/34771 or WO 2005/032582 (including GAS 40), fusions of fragments of GAS M proteins (including those described in WO 02/094851 , and Dale, Vaccine (1999) 17:193-200, and Dale, Vaccine 14(10): 944-948), fibronectin binding protein (Sfb1), Streptococcal heme-associated protein (Shp), and Streptolysin S (SagA).
  • Moraxella catarrhalis: Moraxella antigens include antigens identified in WO 02/18595 and WO 99/58562 , outer membrane protein antigens (HMW-OMP), C-antigen, and/or LPS.
  • Bordetella pertussis: Pertussis antigens include pertussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B. pertussis, optionally also combination with pertactin and/or agglutinogens 2 and 3 antigen.
  • Staphylococcus aureus: Staph aureus antigens include S. aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa exotoxin A, such as StaphVAX™, or antigens derived from surface proteins, invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit phagocytic engulfment (capsule, Protein A), carotenoids, catalase production, Protein A, coagulase, clotting factor, and/or membrane-damaging toxins (optionally detoxified) that lyse eukaryotic cell membranes (hemolysins, leukotoxin, leukocidin).
  • Staphylococcus epidermis: S. epidermidis antigens include slime-associated antigen (SAA).
  • Clostridium tetani (Tetanus): Tetanus antigens include tetanus toxoid (TT), preferably used as a carrier protein in conjunction/conjugated with the compositions of the present invention.
  • Cornynebacterium diphtheriae (Diphtheria): Diphtheria antigens include diphtheria toxin, preferably detoxified, such as CRM197. Additionally antigens capable of modulating, inhibiting or associated with ADP ribosylation are contemplated for combination/coadministration/conjugation with the compositions of the present invention. The diphtheria toxoids may be used as carrier proteins.
  • Haemophilus influenzae B (Hib): Hib antigens include a Hib saccharide antigen.
  • Pseudomonas aeruginosa: Pseudomonas antigens include endotoxin A, Wzz protein, P. aeruginosa LPS, more particularly LPS isolated from PAO1 (O5 serotype), and/or Outer Membrane Proteins, including Outer Membrane Proteins F (OprF) ( Infect Immun. 2001 May; 69(5): 3510-3515).
  • Legionella pneumophila. Bacterial antigens may be derived from Legionella pneumophila.
  • Streptococcus agalactiae (Group B Streptococcus): Group B Streptococcus antigens include a protein or saccharide antigen identified in WO 02/34771 , WO 03/093306 , WO 04/041157 , or WO 2005/002619 (including proteins GBS 80, GBS 104, GBS 276 and GBS 322, and including saccharide antigens derived from serotypes Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII).
  • Neiserria gonorrhoeae: Gonorrhoeae antigens include Por (or porin) protein, such as PorB (see Zhu et al., Vaccine (2004) 22:660 - 669), a transferring binding protein, such as TbpA and TbpB (See Price et al., Infection and Immunity (2004) 71(1):277 - 283), a opacity protein (such as Opa), a reduction-modifiable protein (Rmp), and outer membrane vesicle (OMV) preparations (see Plante et al., J Infectious Disease (2000) 182:848 - 855), also see e.g. WO99/24578 , WO99/36544 , WO99/57280 , WO02/079243 ).
  • Chlamydia trachomatis: Chlamydia trachomatis antigens include antigens derived from serotypes A, B, Ba and C (agents of trachoma, a cause of blindness), serotypes L1, L2 & L3 (associated with Lymphogranuloma venereum), and serotypes, D-K. Chlamydia trachomas antigens may also include an antigen identified in WO 00/37494 , WO 03/049762 , WO 03/068811 , or WO 05/002619 , including PepA (CT045), LcrE (CT089), ArtJ (CT381), DnaK (CT396), CT398, OmpH-like (CT242), L7/L12 (CT316), OmcA (CT444), AtosS (CT467), CT547, Eno (CT587), HrtA (CT823), and MurG (CT761).
  • Treponema pallidum (Syphilis): Syphilis antigens include TmpA antigen.
  • Haemophilus ducreyi (causing chancroid): Ducreyi antigens include outer membrane protein (DsrA).
  • Enterococcus faecalis or Enterococcus faecium: Antigens include a trisaccharide repeat or other Enterococcus derived antigens provided in US Patent No. 6,756,361 .
  • Helicobacter pylori: H pylori antigens include Cag, Vac, Nap, HopX, HopY and/or urease antigen.
  • Staphylococcus saprophyticus: Antigens include the 160 kDa hemagglutinin of S. saprophyticus antigen.
  • Yersinia enterocolitica Antigens include LPS (Infect Immun. 2002 August; 70(8): 4414).
  • E. coli: E. coli antigens may be derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), and/or enterohemorrhagic E. coli (EHEC).
  • Bacillus anthracis (anthrax): B. anthracis antigens are optionally detoxified and may be selected from A-components (lethal factor (LF) and edema factor (EF)), both of which can share a common B-component known as protective antigen (PA).
  • Yersinia pestis (plague): Plague antigens include F1 capsular antigen (Infect Immun. 2003 Jan; 71(1)): 374-383, LPS (Infect Immun. 1999 Oct; 67(10): 5395), Yersinia pestis V antigen (Infect Immun. 1997 Nov; 65(11): 4476-4482).
  • Mycobacterium tuberculosis: Tuberculosis antigens include lipoproteins, LPS, BCG antigens, a fusion protein of antigen 85B (Ag85B) and/or ESAT-6 optionally formulated in cationic lipid vesicles (Infect Immun. 2004 October; 72(10): 6148), Mycobacterium tuberculosis (Mtb) isocitrate dehydrogenase associated antigens (Proc Natl Acad Sci USA. 2004 Aug 24; 101(34):12652), and/or MPT51 antigens (Infect Immun. 2004 July; 72(7): 2004 Aug 24; 101(34): 12652), and/or MPT51 antigens (Infect Immun. 2004 July; 72(7): 3829).
  • Rickettsia: Antigens include outer membrane proteins, including the outer membrane protein A and/or B (OmpB) (Biochim Biophys Acta. 2004 ), LPS, and surface protein antigen (SPA) (J Autoimmun. 1989 Jun;2 Suppl:81).
  • Listeria monocytogenes. Bacterial antigens may be derived from Listeria monocytogenes.
  • Chlamydia pneumoniae: Antigens include those identified in WO 02/02606 .
  • Vibrio cholerae: Antigens include proteinase antigens, LPS, particularly lipopolysaccharides of Vibrio cholerae II, O1 Inaba O-specific polysaccharides, V. cholera 0139, antigens of IEM108 vaccine (Infect Immun. 2003 Oct;71(10):5498-504), and/or Zonula occludens toxin (Zot).
  • Salmonella typhi (typhoid fever): Antigens include capsular polysaccharides preferably conjugates (Vi, i.e. vax-TyVi).
  • Borrelia burgdorferi (Lyme disease): Antigens include lipoproteins (such as OspA, OspB, Osp C and Osp D), other surface proteins such as OspE-related proteins (Erps), decorin-binding proteins (such as DbpA), and antigenically variable VI proteins. , such as antigens associated with P39 and P13 (an integral membrane protein, Infect Immun. 2001 May; 69(5): 3323-3334), VlsE Antigenic Variation Protein (J Clin Microbiol. 1999 Dec; 37(12): 3997).
  • Porphyromonas gingivalis: Antigens include P. gingivalis outer membrane protein (OMP).
  • Klebsiella: Antigens include an OMP, including OMP A, or a polysaccharide optionally conjugated to tetanus toxoid.
  • Further bacterial antigens of the invention may be capsular antigens, polysaccharide antigens or protein antigens of any of the above. Further bacterial antigens may also include an outer membrane vesicle (OMV) preparation. Additionally, antigens include live, attenuated, and/or purified versions of any of the aforementioned bacteria. The antigens of the present invention may be derived from gram-negative or gram-positive bacteria. The antigens of the present invention may be derived from aerobic or anaerobic bacteria.
  • Additionally, any of the above bacterial-derived saccharides (polysaccharides, LPS, LOS or oligosaccharides) can be conjugated to another agent or antigen, such as a carrier protein (for example CRM197). Such conjugation may be direct conjugation effected by reductive amination of carbonyl moieties on the saccharide to amino groups on the protein, as provided in US Patent No. 5,360,897 and Can JBiochem Cell Biol. 1984 May;62(5):270-5. Alternatively, the saccharides can be conjugated through a linker, such as, with succinamide or other linkages provided in Bioconjugate Techniques, 1996 and CRC, Chemistry of Protein Conjugation and Cross-Linking, 1993.
  • B. Viral Antigens
  • Viral antigens suitable for use in the invention include inactivated (or killed) virus, attenuated virus, split virus formulations, purified subunit formulations, viral proteins which may be isolated, purified or derived from a virus, and Virus Like Particles (VLPs). Viral antigens may be derived from viruses propagated on cell culture or other substrate. Alternatively, viral antigens may be expressed recombinantly. Viral antigens preferably include epitopes which are exposed on the surface of the virus during at least one stage of its life cycle. Viral antigens are preferably conserved across multiple serotypes or isolates. Viral antigens include antigens derived from one or more of the viruses set forth below as well as the specific antigens examples identified below.
  • Orthomyxovirus: Viral antigens may be derived from an Orthomyxovirus, such as Influenza A, B and C. Orthomyxovirus antigens may be selected from one or more of the viral proteins, including hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix protein (M1), membrane protein (M2), one or more of the transcriptase components (PB1, PB2 and PA). Preferred antigens include HA and NA.
  • Influenza antigens may be derived from interpandemic (annual) flu strains. Alternatively influenza antigens may be derived from strains with the potential to cause pandemic a pandemic outbreak (i.e., influenza strains with new haemagglutinin compared to the haemagglutinin in currently circulating strains, or influenza strains which are pathogenic in avian subjects and have the potential to be transmitted horizontally in the human population, or influenza strains which are pathogenic to humans).
  • Paramyxoviridae viruses: Viral antigens may be derived from Paramyxoviridae viruses, such as Pneumoviruses (RSV), Paramyxoviruses (PIV) and Morbilliviruses (Measles).
  • Pneumovirus: Viral antigens may be derived from a Pneumovirus, such as Respiratory syncytial virus (RSV), Bovine respiratory syncytial virus, Pneumonia virus of mice, and Turkey rhinotracheitis virus. Preferably, the Pneumovirus is RSV. Pneumovirus antigens may be selected from one or more of the following proteins, including surface proteins Fusion (F), Glycoprotein (G) and Small Hydrophobic protein (SH), matrix proteins M and M2, nucleocapsid proteins N, P and L and nonstructural proteins NS1 and NS2. Preferred Pneumovirus antigens include F, G and M. See e.g., J Gen Virol. 2004 Nov; 85(Pt 11):3229). Pneumovirus antigens may also be formulated in or derived from chimeric viruses. For example, chimeric RSV/PIV viruses may comprise components of both RSV and PIV.
  • Paramyxovirus: Viral antigens may be derived from a Paramyxovirus, such as Parainfluenza virus types 1 - 4 (PIV), Mumps, Sendai viruses, Simian virus 5, Bovine parainfluenza virus and Newcastle disease virus. Preferably, the Paramyxovirus is PIV or Mumps. Paramyxovirus antigens may be selected from one or more of the following proteins: Hemagglutinin -Neuraminidase (HN), Fusion proteins F1 and F2, Nucleoprotein (NP), Phosphoprotein (P), Large protein (L), and Matrix protein (M). Preferred Paramyxovirus proteins include HN, F1 and F2. Paramyxovirus antigens may also be formulated in or derived from chimeric viruses. For example, chimeric RSV/PIV viruses may comprise components of both RSV and PIV. Commercially available mumps vaccines include live attenuated mumps virus, in either a monovalent form or in combination with measles and rubella vaccines (MMR).
  • Morbillivirus: Viral antigens may be derived from a Morbillivirus, such as Measles. Morbillivirus antigens may be selected from one or more of the following proteins: hemagglutinin (H), Glycoprotein (G), Fusion factor (F), Large protein (L), Nucleoprotein (NP), Polymerase phosphoprotein (P), and Matrix (M). Commercially available measles vaccines include live attenuated measles virus, typically in combination with mumps and rubella (MMR).
  • Picornavirus: Viral antigens may be derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and Aphthoviruses. Antigens derived from Enteroviruses, such as Poliovirus are preferred.
  • Enterovirus: Viral antigens may be derived from an Enterovirus, such as Poliovirus types 1, 2 or 3, Coxsackie A virus types 1 to 22 and 24, Coxsackie B virus types 1 to 6, Echovirus (ECHO) virus) types 1 to 9, 11 to 27 and 29 to 34 and Enterovirus 68 to 71. Preferably, the Enterovirus is poliovirus. Enterovirus antigens are preferably selected from one or more of the following Capsid proteins VP1, VP2, VP3 and VP4. Commercially available polio vaccines include Inactivated Polio Vaccine (IPV) and Oral poliovirus vaccine (OPV).
  • Heparnavirus: Viral antigens may be derived from an Heparnavirus, such as Hepatitis A virus (HAV). Commercially available HAV vaccines include inactivated HAV vaccine.
  • Togavirus: Viral antigens may be derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. Antigens derived from Rubivirus, such as Rubella virus, are preferred. Togavirus antigens may be selected from E1, E2, E3, C, NSP-1, NSPO-2, NSP-3 or NSP-4. Togavirus antigens are preferably selected from E1, E2 or E3. Commercially available Rubella vaccines include a live cold-adapted virus, typically in combination with mumps and measles vaccines (MMR).
  • Flavivirus: Viral antigens may be derived from a Flavivirus, such as Tick-borne encephalitis (TBE), Dengue ( types 1, 2, 3 or 4), Yellow Fever, Japanese encephalitis, West Nile encephalitis, St. Louis encephalitis, Russian spring-summer encephalitis, Powassan encephalitis. Flavivirus antigens may be selected from PrM, M, C, E, NS-1, NS-2a, NS2b, NS3, NS4a, NS4b, and NS5. Flavivirus antigens are preferably selected from PrM, M and E. Commercially available TBE vaccine include inactivated virus vaccines.
  • Pestivirus: Viral antigens may be derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).
  • Hepadnavirus: Viral antigens may be derived from a Hepadnavirus, such as Hepatitis B virus. Hepadnavirus antigens may be selected from surface antigens (L, M and S), core antigens (HBc, HBe). Commercially available HBV vaccines include subunit vaccines comprising the surface antigen S protein.
  • Hepatitis C virus: Viral antigens may be derived from a Hepatitis C virus (HCV). (see, e.g. Hsu et al. (1999) Clin Liver Dis 3:901-915). HCV antigens may be selected from one or more of E1, E2, E1/E2, NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions (Houghton et al., Hepatology (1991) 14:381). For example, Hepatitis C virus antigens that may be used can include one or more of the following: HCV E1 and or E2 proteins, E1/E2 heterodimer complexes, core proteins and non-structural proteins, or fragments of these antigens, wherein the non-structural proteins can optionally be modified to remove enzymatic activity but retain immunogenicity (see, e.g. WO03/002065 ; WO01/37869 and WO04/005473 ).
  • Rhabdovirus: Viral antigens may be derived from a Rhabdovirus, such as a Lyssavirus (Rabies virus) and Vesiculovirus (VSV). Rhabdovirus antigens may be selected from glycoprotein (G), nucleoprotein (N), large protein (L), nonstructural proteins (NS). Commercially available Rabies virus vaccine comprise killed virus grown on human diploid cells or fetal rhesus lung cells.
  • Caliciviridae; Viral antigens may be derived from Calciviridae, such as Norwalk virus, and Norwalk-like Viruses, such as Hawaii Virus and Snow Mountain Virus.
  • Coronavirus: Viral antigens may be derived from a Coronavirus, SARS, Human respiratory coronavirus, Avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV). Coronavirus antigens may be selected from spike (S), envelope (E), matrix (M), nucleocapsid (N), and Hemagglutininesterase glycoprotein (HE). Preferably, the Coronavirus antigen is derived from a SARS virus. SARS viral antigens are described in WO 04/92360 ;
  • Retrovirus: Viral antigens may be derived from a Retrovirus, such as an Oncovirus, a Lentivirus or a Spumavirus. Oncovirus antigens may be derived from HTLV-1, HTLV-2 or HTLV-5. Lentivirus antigens may be derived from HIV-1 or HIV-2. Retrovirus antigens may be selected from gag, pol, env, tax, tat, rex, rev, nef, vif, vpu, and vpr. HIV antigens may be selected from gag (p24gag and p55gag), env (gp160 and gp41), pol, tat, nef, rev vpu, miniproteins, (preferably p55 gag and gp140v delete). HIV antigens may be derived from one or more of the following strains: HIVIIIb, HIVSF2, HIVLAV, HIVLAI, HIVMN, HIV-1CM235, HIV-1US4.
  • Reovirus: Viral antigens may be derived from a Reovirus, such as an Orthoreovirus, a Rotavirus, an Orbivirus, or a Coltivirus. Reovirus antigens may be selected from structural proteins λ1, λ2, λ3, µ1, µ2, σ1, σ2, or σ3, or nonstructural proteins σNS, µNS, or σ1s. Preferred Reovirus antigens may be derived from a Rotavirus. Rotavirus antigens may be selected from VP1, VP2, VP3, VP4 (or the cleaved product VP5 and VP8), NSP 1, VP6, NSP3, NSP2, VP7, NSP4, or NSP5. Preferred Rotavirus antigens include VP4 (or the cleaved product VP5 and VP8), and VP7.
  • Parvovirus: Viral antigens may be derived from a Parvovirus, such as Parvovirus B19. Parvovirus antigens may be selected from VP-1, VP-2, VP-3, NS-1 and NS-2. Preferably, the Parvovirus antigen is capsid protein VP-2.
  • Delta hepatitis virus (HDV): Viral antigens may be derived HDV, particularly δ-antigen from HDV (see, e.g., U.S. Patent No. 5,378,814 ).
  • Hepatitis E virus (HEV): Viral antigens may be derived from HEV.
  • Hepatitis G virus (HGV): Viral antigens may be derived from HGV.
  • Human Herpesvirus: Viral antigens may be derived from a Human Herpesvirus, such as Herpes Simplex Viruses (HSV), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human Herpesvirus 8 (HHV8). Human Herpesvirus antigens may be selected from immediate early proteins (α), early proteins (β), and late proteins (γ). HSV antigens may be derived from HSV-1 or HSV-2 strains. HSV antigens may be selected from glycoproteins gB, gC, gD and gH, fusion protein (gB), or immune escape proteins (gC, gE, or gI). VZV antigens may be selected from core, nucleocapsid, tegument, or envelope proteins. A live attenuated VZV vaccine is commercially available. EBV antigens may be selected from early antigen (EA) proteins, viral capsid antigen (VCA), and glycoproteins of the membrane antigen (MA). CMV antigens may be selected from capsid proteins, envelope glycoproteins (such as gB and gH), and tegument proteins
  • Papovaviruses: Antigens may be derived from Papovaviruses, such as Papillomaviruses and Polyomaviruses. Papillomaviruses include HPV serotypes 1, 2, 4, 5, 6, 8, 11, 13, 16, 18, 31, 33, 35, 39, 41, 42, 47, 51, 57, 58, 63 and 65. Preferably, HPV antigens are derived from serotypes 6, 11, 16 or 18. HPV antigens may be selected from capsid proteins (L1) and (L2), or E1 - E7, or fusions thereof. HPV antigens are preferably formulated into virus-like particles (VLPs). Polyomyavirus viruses include BK virus and JK virus. Polyomavirus antigens may be selected from VP1, VP2 or VP3.
  • Further provided are antigens, compositions, methods, and microbes included in Vaccines, 4th Edition (Plotkin and Orenstein ed. 2004); Medical Microbiology 4th Edition (Murray et al. ed. 2002); Virology, 3rd Edition (W.K. Joklik ed. 1988); Fundamental Virology, 2nd Edition (B.N. Fields and D.M. Knipe, eds. 1991), which are contemplated in conjunction with the compositions of the present invention.
  • C. Fungal Antigens
  • Fungal antigens for use in the invention may be derived from one or more of the fungi set forth below.
  • Fungal antigens may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var. discoides, var. ochraceum, Trichophyton violaceum, and/or Trichophyton faviforme.
  • Fungal pathogens may be derived from Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida parapsilosis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladosporium carrionii, Coccidioides immitis, Blastomyces dermatidis, Cryptococcus neoformans, Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Paracoccidioides brasiliensis, Pneumocystis carinii, Pythiumn insidiosum, Pityrosporum ovale, Sacharomyces cerevisae, Saccharomyces boulardii, Saccharomyces pombe, Scedosporium apiosperum, Sporothrix schenckii, Trichosporon beigelii, Toxoplasma gondii, Penicillium marneffei, Malassezia spp., Fonsecaea spp., Wangiella spp., Sporothrix spp., Basidiobolus spp., Conidiobolus spp., Rhizopus spp, Mucor spp, Absidia spp, Mortierella spp, Cunninghamella spp, Saksenaea spp., Alternaria spp, Curvularia spp, Helminthosporium spp, Fusarium spp, Aspergillus spp, Penicillium spp, Monolinia spp, Rhizoctonia spp, Paecilomyces spp, Pithomyces spp, and Cladosporium spp.
  • Processes for producing a fungal antigens are well known in the art (see US Patent No. 6,333,164 ). In a preferred method a solubilized fraction extracted and separated from an insoluble fraction obtainable from fungal cells of which cell wall has been substantially removed or at least partially removed, characterized in that the process comprises the steps of: obtaining living fungal cells; obtaining fungal cells of which cell wall has been substantially removed or at least partially removed; bursting the fungal cells of which cell wall has been substantially removed or at least partially removed; obtaining an insoluble fraction; and extracting and separating a solubilized fraction from the insoluble fraction.
  • D. STD Antigens
  • The compositions of the invention may include one or more antigens derived from a sexually transmitted disease (STD). Such antigens may provide for prophylactis or therapy for STD's such as chlamydia, genital herpes, hepatits (such as HCV), genital warts, gonorrhoea, syphilis and/or chancroid (See, WO00/15255 ). Antigens may be derived from one or more viral or bacterial STD's. Viral STD antigens for use in the invention may be derived from, for example, HIV, herpes simplex virus (HSV-1 and HSV-2), human papillomavirus (HPV), and hepatitis (HCV). Bacterial STD antigens for use in the invention may be derived from, for example, Neiserria gonorrhoeae, Chlamydia trachomatis, Treponema pallidum, Haemophilus ducreyi, E. coli, and Streptococcus agalactiae. Examples of specific antigens derived from these pathogens are described above.
  • E. Respiratory Antigens
  • The compositions of the invention may include one or more antigens derived from a pathogen which causes respiratory disease. For example, respiratory antigens may be derived from a respiratory virus such as Orthomyxoviruses (influenza), Pneumovirus (RSV), Paramyxovirus (PIV), Morbillivirus (measles), Togavirus (Rubella), VZV, and Coronavirus (SARS). Respiratory antigens may be derived from a bacteria which causes respiratory disease, such as Streptococcus pneumoniae, Pseudomonas aeruginosa, Bordetella pertussis, Mycobacterium tuberculosis, Mycoplasma pneumoniae, Chlamydia pneumoniae, Bacillus anthracis, and Moraxella catarrhalis. Examples of specific antigens derived from these pathogens are described above.
  • F. Pediatric Vaccine Antigens
  • The compositions of the invention may include one or more antigens suitable for use in pediatric subjects. Pediatric subjects are typically less than about 3 years old, or less than about 2 years old, or less than about 1 years old. Pediatric antigens may be administered multiple times over the course of 6 months, 1, 2 or 3 years. Pediatric antigens may be derived from a virus which may target pediatric populations and/or a virus from which pediatric populations are susceptible to infection. Pediatric viral antigens include antigens derived from one or more of Orthomyxovirus (influenza), Pneumovirus (RSV), Paramyxovirus (PIV and Mumps), Morbillivirus (measles), Togavirus (Rubella), Enterovirus (polio), HBV, Coronavirus (SARS), and Varicella-zoster virus (VZV), Epstein Barr virus (EBV). Pediatric bacterial antigens include antigens derived from one or more of Streptococcus pneumoniae, Neisseria meningitides, Streptococcus pyogenes (Group A Streptococcus), Moraxella catarrhalis, Bordetella pertussis, Staphylococcus aureus, Clostridium tetani (Tetanus), Cornynebacterium diphtheriae (Diphtheria), Haemophilus influenzae B (Hib), Pseudomonas aeruginosa, Streptococcus agalactiae (Group B Streptococcus), and E. coli. Examples of specific antigens derived from these pathogens are described above.
  • G. Antigens suitable for use in Elderly or Immunocompromised Individuals
  • The compositions of the invention may include one or more antigens suitable for use in elderly or immunocompromised individuals. Such individuals may need to be vaccinated more frequently, with higher doses or with adjuvanted formulations to improve their immune response to the targeted antigens. Antigens which may be targeted for use in Elderly or Immunocompromised individuals include antigens derived from one or more of the following pathogens: Neisseria meningitides, Streptococcus pneumoniae, Streptococcus pyogenes (Group A Streptococcus), Moraxella catarrhalis, Bordetella pertussis, Staphylococcus aureus, Staphylococcus epidermis, Clostridium tetani (Tetanus), Cornynebacterium diphtheriae (Diphtheria), Haemophilus influenzae B (Hib), Pseudomonas aeruginosa, Legionella pneumophila, Streptococcus agalactiae (Group B Streptococcus), Enterococcus faecalis, Helicobacter pylori, Clamydia pneumoniae, Orthomyxovirus (influenza), Pneumovirus (RSV), Paramyxovirus (PIV and Mumps), Morbillivirus (measles), Togavirus (Rubella), Enterovirus (polio), HBV, Coronavirus (SARS), Varicella-zoster virus (VZV), Epstein Barr virus (EBV), Cytomegalovirus (CMV). Examples of specific antigens derived from these pathogens are described above.
  • H. Antigens suitable for use in Adolescent Vaccines
  • The compositions of the invention may include one or more antigens suitable for use in adolescent subjects. Adolescents may be in need of a boost of a previously administered pediatric antigen. Pediatric antigens which may be suitable for use in adolescents are described above. In addition, adolescents may be targeted to receive antigens derived from an STD pathogen in order to ensure protective or therapeutic immunity before the beginning of sexual activity. STD antigens which may be suitable for use in adolescents are described above.
  • I. Tumor Antigens
  • One embodiment of the present involves a tumor antigen or cancer antigen in conjunction with the compositions of the present invention. Tumor antigens can be, for example, peptide-containing tumor antigens, such as a polypeptide tumor antigen or glycoprotein tumor antigens. A tumor antigen can also be, for example, a saccharide-containing tumor antigen, such as a glycolipid tumor antigen or a ganglioside tumor antigen. The tumor antigen can further be, for example, a polynucleotide-containing tumor antigen that expresses a polypeptide-containing tumor antigen, for instance, an RNA vector construct or a DNA vector construct, such as plasmid DNA.
  • Tumor antigens appropriate for the practice of the present invention encompass a wide variety of molecules, such as (a) polypeptide-containing tumor antigens, including polypeptides (which can range, for example, from 8-20 amino acids in length, although lengths outside this range are also common), lipopolypeptides and glycoproteins, (b) saccharide-containing tumor antigens, including poly-saccharides, mucins, gangliosides, glycolipids and glycoproteins, and (c) polynucleotides that express antigenic polypeptides.
  • The tumor antigens can be, for example, (a) full length molecules associated with cancer cells, (b) homologs and modified forms of the same, including molecules with deleted, added and/or substituted portions, and (c) fragments of the same. Tumor antigens can be provided in recombinant form. Tumor antigens include, for example, class I-restricted antigens recognized by CD8+ lymphocytes or class II-restricted antigens recognized by CD4+ lymphocytes.
  • Numerous tumor antigens are known in the art, including: (a) cancer-testis antigens such as NY-ESO-1, SSX2, SCP1 as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-1, GAGE-2, MAGE-1, MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors), (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUM1 (associated with, e.g., melanoma), caspase-8 (associated with, e.g., head and neck cancer), CIA 0205 (associated with, e.g., bladder cancer), HLA-A2-R1701, beta catenin (associated with, e.g., melanoma), TCR (associated with, e.g., T-cell non-Hodgkins lymphoma), BCR-abl (associated with, e.g., chronic myelogenous leukemia), triosephosphate isomerase, KIA 0205, CDC-27, and LDLR-FUT, (c) over-expressed antigens, for example, Galectin 4 (associated with, e.g., colorectal cancer), Galectin 9 (associated with, e.g., Hodgkin's disease), proteinase 3 (associated with, e.g., chronic myelogenous leukemia), WT 1 (associated with, e.g., various leukemias), carbonic anhydrase (associated with, e.g., renal cancer), aldolase A (associated with, e.g., lung cancer), PRAME (associated with, e.g., melanoma), HER-2/neu (associated with, e.g., breast, colon, lung and ovarian cancer), alpha-fetoprotein (associated with, e.g., hepatoma), KSA (associated with, e.g., colorectal cancer), gastrin (associated with, e.g., pancreatic and gastric cancer), telomerase catalytic protein, MUC-1 (associated with, e.g., breast and ovarian cancer), G-250 (associated with, e.g., renal cell carcinoma), p53 (associated with, e.g., breast, colon cancer), and carcinoembryonic antigen (associated with, e.g., breast cancer, lung cancer, and cancers of the gastrointestinal tract such as colorectal cancer), (d) shared antigens, for example, melanoma-melanocyte differentiation antigens such as MART-1/Melan A, gp100, MC1R, melanocyte-stimulating hormone receptor, tyrosinase, tyrosinase related protein-1/TRP1 and tyrosinase related protein-2/TRP2 (associated with, e.g., melanoma), (e) prostate associated antigens such as PAP, PSA, PSMA, PSH-P1, PSM-P1, PSM-P2, associated with e.g., prostate cancer, (f) immunoglobulin idiotypes (associated with myeloma and B cell lymphomas, for example), and (g) other tumor antigens, such as polypeptide- and saccharide-containing antigens including (i) glycoproteins such as sialyl Tn and sialyl Lex (associated with, e.g., breast and colorectal cancer) as well as various mucins; glycoproteins may be coupled to a carrier protein (e.g., MUC-1 may be coupled to KLH); (ii) lipopolypeptides (e.g., MUC-1 linked to a lipid moiety); (iii) polysaccharides (e.g., Globo H synthetic hexasaccharide), which may be coupled to a carrier proteins (e.g., to KLH), (iv) gangliosides such as GM2, GM12, GD2, GD3 (associated with, e.g., brain, lung cancer, melanoma), which also may be coupled to carrier proteins (e.g., KLH). Additional tumor antigens which are known in the art include p15, Hom/Mel-40, H-Ras, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, p185erbB2, p180erbB-3, c-met, mn-23H1, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, p16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29\BCAA), CA 195, CA 242, CA-50, CAM43, CD68\KP1, CO-029, FGF-5, Ga733 (EpCAM), HTgp-175, M344, MA-50, MG7-Ag, MOV18, NB/70K, NY-CO-1, RCAS1, SDCCAG16, TA-90 (Mac-2 binding protein\cyclophilin C-associated protein), TAAL6, TAG72, TLP, TPS, and the like. These as well as other cellular components are described for example in United States Patent Application 20020007173 and references cited therein.
  • Polynucleotide-containing antigens in accordance with the present invention typically comprise polynucleotides that encode polypeptide cancer antigens such as those listed above. Preferred polynucleotide-containing antigens include DNA or RNA vector constructs, such as plasmid vectors (e.g., pCMV), which are capable of expressing polypeptide cancer antigens in vivo.
  • Tumor antigens may be derived, for example, from mutated or altered cellular components. After alteration, the cellular components no longer perform their regulatory functions, and hence the cell may experience uncontrolled growth. Representative examples of altered cellular components include ras, p53, Rb, altered protein encoded by the Wilms' tumor gene, ubiquitin, mucin, protein encoded by the DCC, APC, and MCC genes, as well as receptors or receptor-like structures such as neu, thyroid hormone receptor, platelet derived growth factor (PDGF) receptor, insulin receptor, epidermal growth factor (EGF) receptor, and the colony stimulating factor (CSF) receptor. These as well as other cellular components are described for example in U.S. Patent No. 5,693,522 and references cited therein.
  • Additionally, bacterial and viral antigens, may be used in conjunction with the compositions of the present invention for the treatment of cancer. In particular, carrier proteins, such as CRM197, tetanus toxoid, or Salmonella typhimurium antigen can be used in conjunction/conjugation with compounds of the present invention for treatment of cancer. The cancer antigen combination therapies will show increased efficacy and bioavailability as compared with existing therapies.
  • Additional information on cancer or tumor antigens can be found, for example, in Moingeon P, "Cancer vaccines," Vaccine, 2001, 19:1305-1326; Rosenberg SA, "Progress in human tumor immunology and immunotherapy," Nature, 2001, 411:380-384; Dermine, S. et al, "Cancer Vaccines and Immunotherapy," British Medical Bulletin, 2002, 62, 149-162; Espinoza-Delgado I., "Cancer Vaccines," The Oncologist, 2002, 7(suppl3):20-33; Davis, I.D. et al., "Rational approaches to human cancer immunotherapy," Journal of Leukocyte Biology, 2003, 23: 3-29; Van den Eynde B, et al., "New tumor antigens recognized by T cells," Curr. Opin. Immunol., 1995, 7:674-81; Rosenberg SA, "Cancer vaccines based on the identification of genes encoding cancer regression antigens, Immunol. Today, 1997, 18:175-82; Offringa R et al., "Design and evaluation of antigen-specific vaccination strategies against cancer," Current Opin. Immunol., 2000, 2:576-582; Rosenberg SA, "A new era for cancer immunotherapy based on the genes that encode cancer antigens," Immunity, 1999, 10:281-7; Sahin U et al., "Serological identification of human tumor antigens," Curr. Opin. Immunol., 1997, 9:709-16; Old LJ et al., "New paths in human cancer serology," J. Exp. Med., 1998, 187:1163-7; Chaux P, et al., "Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes," J. Exp. Med., 1999, 189:767-78; Gold P, et al., "Specific carcinoembryonic antigens of the human digestive system," J. Exp. Med., 1965, 122:467-8; Livingston PO, et al., Carbohydrate vaccines that induce antibodies against cancer: Rationale," Cancer Immunol. Immunother., 1997, 45:1-6; Livingston PO, et al., Carbohydrate vaccines that induce antibodies against cancer: Previous experience and future plans," Cancer Immunol. Immunother., 1997, 45:10-9; Taylor-Papadimitriou J, "Biology, biochemistry and immunology of carcinomaassociated mucins," Immunol. Today, 1997, 18:105-7; Zhao X-J et al., "GD2 oligosaccharide: target for cytotoxic T lymphocytes," J. Exp. Med., 1995, 182:67-74; Theobald M, et al., "Targeting p53 as a general tumor antigen," Proc. Natl. Acad. Sci. USA, 1995, 92:11993-7; Gaudernack G, "T cell responses against mutant ras: a basis for novel cancer vaccines," Immunotechnology, 1996, 2:3-9; WO 91/02062 ; U.S. Patent No. 6,015,567 ; WO 01/08636 ; WO 96/30514 ; U.S. Patent No. 5,846,538 ; and U.S. Patent No. 5,869,445 .
  • Antigen Formulations
  • In other aspects of the invention, methods of producing microparticles having adsorbed antigens are provided. The methods comprise: (a) providing an emulsion by dispersing a mixture comprising (i) water, (ii) a detergent, (iii) an organic solvent, and (iv) a biodegradable polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate. The polymer is typically present in the mixture at a concentration of about 1% to about 30% relative to the organic solvent, while the detergent is typically present in the mixture at a weight-to-weight detergent-to-polymer ratio of from about 0.00001:1 to about 0.1:1 (more typically about 0.0001:1 to about 0.1:1, about 0.001:1 to about 0.1:1, or about 0.005:1 to about 0.1:1); (b) removing the organic solvent from the emulsion; and (c) adsorbing an antigen on the surface of the microparticles. In certain embodiments, the biodegradable polymer is present at a concentration of about 3% to about 10% relative to the organic solvent.
  • Microparticles for use herein will be formed from materials that are sterilizable, non-toxic and biodegradable. Such materials include, without limitation, poly(α-hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate. Preferably, microparticles for use with the present invention are derived from a poly(α-hydroxy acid), in particular, from a poly(lactide) ("PLA") or a copolymer of D,L-lactide and glycolide or glycolic acid, such as a poly(D,L-lactide-co-glycolide) ("PLG" or "PLGA"), or a copolymer of D,L-lactide and caprolactone. The microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of which will be largely a matter of choice, depending in part on the coadministered macromolecule. These parameters are discussed more fully below.
  • Further antigens may also include an outer membrane vesicle (OMV) preparation.
  • Additional formulation methods and antigens (especially tumor antigens) are provided in U.S. Patent Serial No. 09/581,772 .
  • Pharmaceutical compositions that include the compounds described herein may include additives such as excipients. Suitable pharmaceutically acceptable excipients include processing agents and drug delivery modifiers and enhancers, such as, for example, calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, dextrose, hydroxypropyl-β-cyclodextrin, polyvinylpyrrolidinone, low melting waxes, ion exchange resins, and the like, as well as combinations of any two or more of these. Other suitable pharmaceutically acceptable excipients are described in "Remington's Pharmaceutical Sciences," Mack Pub. Co., New Jersey (1991), which is hereby incorporated herein by reference in its entirety and for all purposes as if fully set forth herein.
  • Pharmaceutical compositions that include the compounds of the invention may be in any form suitable for the intended method of administration, including, for example, as a solution, a suspension, or an emulsion. Liquid carriers are typically used in preparing solutions, suspensions, and emulsions. Liquid carriers contemplated for use in the practice of the present invention include, for example, water, saline, pharmaceutically acceptable organic solvent(s), pharmaceutically acceptable oils or fats, and the like, as well as mixtures of two or more of these. The liquid carrier may include other suitable pharmaceutically acceptable additives such as solubilizers, emulsifiers, nutrients, buffers, preservatives, suspending agents, thickening agents, viscosity regulators, stabilizers, and the like. Suitable organic solvents include, for example, monohydric alcohols, such as ethanol, and polyhydric alcohols, such as glycols. Suitable oils include, but are not limited to, soybean oil, coconut oil, olive oil, safflower oil, cottonseed oil, and the like. For parenteral administration, the carrier may be an oily ester such as ethyl oleate, isopropyl myristate, and the like. Compositions of the present invention may also be in the form of microparticles, microcapsules, and the like, as well as combinations of any two or more of these.
  • The compounds and combinations of the present invention can also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form may include, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like. Preferred lipids include phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods of forming liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.W., p. 33 et seq (1976).
  • Controlled release delivery systems may also be used, such as a diffusion controlled matrix system or an erodible system, as described for example in: Lee, "Diffusion-Controlled Matrix Systems", pp. 155-198 and Ron and Langer, "Erodible Systems", pp. 199-224, in "Treatise on Controlled Drug Delivery", A. Kydonieus Ed., Marcel Dekker, Inc., New York 1992. The matrix may be, for example, a biodegradable material that can degrade spontaneously in situ and in vivo for, example, by hydrolysis or enzymatic cleavage, e.g., by proteases. The delivery system may be, for example, a naturally occurring or synthetic polymer or copolymer, for example in the form of a hydrogel. Exemplary polymers with cleavable linkages include polyesters, polyorthoesters, polyanhydrides, polysaccharides, poly(phosphoesters), polyamides, polyurethanes, poly(imidocarbonates) and poly(phosphazenes).
  • The compounds of the invention may be administered enterally, orally, parenterally, sublingually, intradermally, by inhalation spray, rectally, or topically in dosage unit formulations that include conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. For example, suitable modes of administration include oral, subcutaneous, transdermal, transmucosal, iontophoretic, intravenous, intramuscular, intraperitoneal, intranasal, subdermal, rectal, and the like. Topical administration may also include the use of transdermal administration such as transdermal patches or ionophoresis devices. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques.
  • Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-propanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch. Such dosage forms may also include, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also include buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, cyclodextrins, and sweetening, flavoring, and perfuming agents.
  • Effective amounts of the compounds of the invention generally include any amount sufficient to detectably treat the disorders described herein.
  • Successful treatment of a subject in accordance with the invention may result in a reduction or alleviation of symptoms in a subject afflicted with a medical or biological disorder. For example, treatment may halt the further progression of the disorder, or may prevent or retard development of the disorder.
  • The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and severity of the particular disease undergoing therapy. The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
  • Antigen References
  • The following references include antigens useful in conjunction with the compositions of the present invention:
    • Antigen references are listed below:
      1. 1. International patent application WO 99/24578
      2. 2. International patent application WO 99/36544 .
      3. 3. International patent application WO 99/57280 .
      4. 4. International patent application WO 00/22430 .
      5. 5. Tettelin et al. (2000) Science 287:1809-1815.
      6. 6. International patent application WO 96/29412 .
      7. 7. Pizza et al. (2000) Science 287:1816-1820.
      8. 8. PCT WO 01/52885 .
      9. 9. Bjune et al. (1991) Lancet 338(8775).
      10. 10. Fuskasawa et al. (1999) Vaccine 17:2951-2958.
      11. 11. Rosenqist et al. (1998) Dev. Biol. Strand 92:323-333.
      12. 12. Constantino et al. (1992) Vaccine 10:691-698.
      13. 13. Constantino et al. (1999) Vaccine 17:1251-1263.
      14. 14. Watson (2000) Pediatr Infect Dis J 19:331-332.
      15. 15. Rubin (20000) Pediatr Clin North Am 47:269-285,v.
      16. 16. Jedrzejas (2001) Microbiol Mol Biol Rev 65:187-207.
      17. 17. International patent application filed on 3rd July 2001 claiming priority from GB-0016363.4 ; WO 02/02606 ; PCT IB/01/00166 .
      18. 18. Kalman et al. (1999) Nature Genetics 21:385-389.
      19. 19. Read et al. (2000) Nucleic Acids Res 28:1397-406.
      20. 20. Shirai et al. (2000) J. Infect. Dis 181(Suppl 3):S524-S527.
      21. 21. International patent application WO 99/27105 .
      22. 22. International patent application WO 00/27994 .
      23. 23. International patent application WO 00/37494 .
      24. 24. International patent application WO 99/28475 .
      25. 25. Bell (2000) Pediatr Infect Dis J 19:1187-1188.
      26. 26. Iwarson (1995) APMIS 103:321-326.
      27. 27. Gerlich et al. (1990) Vaccine 8 Suppl:S63-68 & 79-80.
      28. 28. Hsu et al. (1999) Clin Liver Dis 3:901-915.
      29. 29. Gastofsson et al. (1996) N. Engl. J. Med. 334-:349-355.
      30. 30. Rappuoli et al. (1991) TIBTECH 9:232-238.
      31. 31. Vaccines (1988) eds. Plotkin & Mortimer. ISBN 0-7216-1946-0.
      32. 32. Del Guidice et al. (1998) Molecular Aspects of Medicine 19:1-70.
      33. 33. International patent application WO 93/018150 .
      34. 34. International patent application WO 99/53310 .
      35. 35. International patent application WO 98/04702 .
      36. 36. Ross et al. (2001) Vaccine 19:135-142.
      37. 37. Sutter et al. (2000) Pediatr Clin North Am 47:287-308.
      38. 38. Zimmerman & Spann (1999) Am Fan Physician 59:113-118, 125-126.
      39. 39. Dreensen (1997) Vaccine 15 Suppl"S2-6.
      40. 40. MMWR Morb Mortal Wkly rep 1998 Jan 16:47(1):12, 9.
      41. 41. McMichael (2000) Vaccinel9 Suppl 1:S101-107.
      42. 42. Schuchat (1999) Lancer 353(9146):51-6.
      43. 43. GB patent applications 0026333.5 , 0028727.6 & 0105640.7 .
      44. 44. Dale (1999) Infect Disclin North Am 13:227-43, viii.
      45. 45. Ferretti et al. (2001) PNAS USA 98: 4658-4663.
      46. 46. Kuroda et al. (2001) Lancet 357(9264):1225-1240; see also pages 1218-1219.
      47. 47. Ramsay et al. (2001) Lancet 357(9251):195-196.
      48. 48. Lindberg (1999) Vaccine 17 Suppl 2:S28-36.
      49. 49. Buttery & Moxon (2000) J R Coil Physicians Long 34:163-168.
      50. 50. Ahmad & Chapnick (1999) Infect Dis Clin North Am 13:113-133, vii.
      51. 51. Goldblatt (1998) J. Med. Microbiol. 47:663-567.
      52. 52. European patent 0 477 508 .
      53. 53. U.S. Patent No. 5,306,492 .
      54. 54. International patent application WO 98/42721 .
      55. 55. Conjugate Vaccines (eds. Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114.
      56. 56. Hermanson (1996) Bioconjugate Techniques ISBN: 012323368 & 012342335X.
      57. 57. European patent application 0372501 .
      58. 58. European patent application 0378881 .
      59. 59. European patent application 0427347 .
      60. 60. International patent application WO 93/17712 .
      61. 61. International patent application WO 98/58668 .
      62. 62. European patent application 0471177 .
      63. 63. International patent application WO 00/56360 .
      64. 64. International patent application WO 00/67161 .
    DEFINITIONS
  • As used above and elsewhere herein the following terms and abbreviations have the meanings defined below:
  • AcH
    Acetic Acid
    ATP
    Adenosine triphosphate
    BCG
    Mycobacterium bovis bacillus Calmette-Guerin
    Bn
    Benzyl
    BSA
    Bovine Serum Albumin
    DCM
    Dichloromethane
    DIEA
    N,N-diisopropyl-ethylamine
    EDC
    1-(3-Dimethylaminopropyl)3-ethylcarbodiimide
    hydrochloride FHA
    Filamentous haemaglutinin
    GCMS
    Gas Chromatography / Mass Spectroscopy
    H. Pylori
    Helicobacter Pylori
    HAV
    Hepatitis A Virus
    HBV
    Hepatitis B Virus
    HBr
    Hydrogen Bromide
    HCV
    Hepatitis C Virus
    HIV
    Human Immunodeficiency Virus
    HPLC
    High Performance Liquid Chromatography
    HSV
    Herpes Simplex Virus
    IC50 value
    The concentration of an inhibitor that causes a 50 % reduction in a measured activity.
    IFN
    Interferon
    IL
    Interleukin
    IMS
    Immunomagnetic separation
    IPV
    Inactivated polio virus
    LCMS
    Liquid Chromatography / Mass Spectroscopy
    LPS
    Lipid polysaccharide
    MAb or mAb
    Monoclonal Antibody
    Men A
    Neisseria Meningitidis Type A
    Men C
    Neisseria Meningitidis Type C
    Men B
    Neisseria Meningitidis Type B
    Men W
    Neisseria Meningitidis Type W
    Men Y
    Neisseria Meningitidis Type Y
    MeOH
    Methanol
    MW
    Molecular Weight
    NANB
    Non-A, non-B hepatitis
    NMR
    Nuclear magnetic resonance
    OMV
    Outer membrane vesicle
    PBMC
    Peripheral blood mononuclear cells
    PT
    Pertussis holotoxin
    Rt
    Room temperature (25°C)
    SMIP
    Small Molecule Immune Potentiator
    tBOK
    Potassium Tertiary Butoxide
    TEA
    Triethylamine
    OTf
    Triflate
    THF
    Tetrahydrofuran
    TLC
    Thin Layer Chromatography and/or Tender Loving Care
    TMS
    Trimethylsilyl
    TNF-α
    Tumour necrosis factor-alpha
  • The term "SMIP" refers to a small molecule immunopotentiating compound, including small molecule compounds, generally below about MW 800 g/mol, capable of stimulating or modulating a pro-inflammatory response in a patient. In some embodiments, the SMIP compounds are able to stimulate human peripheral blood mononuclear cells to produce cytokines. More particularly, preferred SMIPs include imidazoquinolines and those compounds encompassed by Formulas (I)a nd (II) described herein, or contained within any reference cited herein.
  • The term "SMIS" refers to a small molecule immunosuppressant compound, including small molecule compounds, generally below about MW 800 g/mol, capable of suppressing or modulating an immune response in a patient. In some embodiments, the SMIS compounds are able to inhibit human peripheral blood mononuclear cell's ability to produce cytokines, chemokines, and/or growth factors. In other embodiments, the SMIS compounds are able to induce TGF-beta production, thereby suppressing an immune response.
  • Reference to "imidazoquinoxalines" (as pertaining to imidazoquinoxalincs of the present invention), indicates compounds imidazo[1,2-a]quinoxalines having the general structure of Formula (I) or (II) as described herein. Some preferred imidazoquinoxalines include compound of Formula:
    Figure imgb0031
  • The term "refractory cancer cells" refers to cancer cell lines that are resistant to preexisting therapeutics or treatment regimens, including prescribed dosing schedules.
  • The methods of the invention are useful in treating "allergic diseases," which may be accomplished in the same manner as the other immunotherapeutic methods described herein.
  • An "allergen" refers to a substance (antigen) that can induce an allergic or asthmatic response in a susceptible subject. The list of allergens is enormous and can include pollens, insect venoms, animal dander, dust, fungal spores, and drugs (e.g., penicillin).
  • "Asthma" refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways, and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively, associated with atopic or allergic symptoms.
  • The term "leukotriene inhibitor" includes any agent or compound that inhibits, restrains, retards, or otherwise interacts with the action or activity of leukotrienes, such as, but not limited to, 5-lipoxygenase ("5-LO") inhibitors, 5-lipoxygenase activating protein ("FLAP") antagonists, and leukotriene D4 ("LTD4 ") antagonists.
  • "Modulating" refers to inducing or suppressing.
  • "Immune-stimulation" "immune response" or "immune potentiation" refer to enhancement or activation of the immune system, including humoral or cellular activation, for example, activation of a cell, such as a killer (T or NK) or dendritic cell of the immune system, for example, causing the increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • "Modulating an immune response" refers to either immune potentiation or immune suppression as defined herein.
  • An "immunogenic composition" refers to a composition capable of stimulating an immune response. In some embodiments, "immunogenic compositions" are compositions capable of stimulating an immune response in a subject. In some embodiments, the immunogenic composition is capable of modulating the production of cytokines in a subject, thereby effecting immune potentiation in that subject.
  • "Immune suppression" or "immunosuppression" refers to deactivation of the immune system, for example, preventing or lessening cytokine production from a dendritic cell leading to an overall attenuation of host defense (immune response).
  • An "immune-stimulatory effective amount" is an amount effective for activating the immune system, for example, causing an increase in cytokine production from a dendritic cell leading to an overall enhancement of host defense (immune response).
  • "Enhancing the immune response to an antigen" by a compound refers to enhancement of the immune response in comparison to that in the absence of the compound. An enhanced immune-response eliciting composition is a composition generally comprising an antigen and a small molecule immune potentiator compound that elicits an immune response greater than a composition comprising an antigen and not containing one or more small molecule immune potentiator compounds. In such embodiments, the compound acts as an adjuvant, for example, for use in vaccine compositions and methods.
  • A "disease associated with cellular proliferation" includes, but is not limited to ncuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis, proliferative diabetic retinopathy (PDR), hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, angiogcncsis, and endotoxic shock.
  • The term "effective amount" is an amount necessary or sufficient to realize a desired biological effect. For example, an effective amount of a compound to treat an infectious disorder may be an amount necessary to cause an antigen specific immune response upon exposure to an infectious agent. The effective amount may vary, depending, for example, upon the condition treated, weight of the subject and severity of the disease. One of skill in the art can readily determine the effective amount empirically without undue experimentation.
  • As used herein "an effective amount for treatment" refers to an amount sufficient to palliate, ameliorate, stabilize, reverse, slow or delay progression of a condition such as a disease state.
  • Reference to "metronomic administration" or "administered metronomically" refers to increasingly frequent dosing regimens, at lower drug concentrations, as compared with known dosing regimens for an existing therapeutic. Metronomic administration varies from the typical dosing of cytotoxic drugs, which involves episodic (less frequent) administration at maximum tolerated doses (MTDs).
  • A "subject" or "patient" is meant to describe a human or vertebrate animal including a dog, cat, pocket pet, marmoset, horse, cow, pig, sheep, goat, elephant, giraffe, chicken, lion, monkey, owl, rat, squirrel, slender loris, and mouse.
  • A "pocket pet" refers to a group of vertebrate animals capable of fitting into a commodious coat pocket such as, for example, hamsters, chinchillas, ferrets, rats, guinea pigs, gerbils, rabbits and sugar gliders. Further description is provided by Mackay, B., Pocket Pets, Animal Issues, 32(1) 2001.
  • As used herein, the term "pharmaceutically acceptable ester" refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkancdioic acids, in which each alkyl or alkenyl moicty advantageously has not more than 6 carbon atoms. Representative examples of particular esters include, but arc not limited to, formates, acetates, propionates, butyrates, acrylatcs and ethylsuccinates.
  • The compounds of the present invention can be used in the form of salts as in "pharmaceutically acceptable salts" derived from inorganic or organic acids. These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-napthalenesulfonate, oxalate, pamoate, pectinate, sulfate, 3-phenylpropionate, picratc, pivalatc, propionate, succinate, tartratc, thiocyanate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • The term "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987. Prodrugs as described in U.S. Patent No. 6,284,772 for example may be used.
  • The symbol
    Figure imgb0032
    is meant to indicate the point of attachment of an appendage.
  • Reference to "halo," "halide," or "halogen" refers to F, Cl, Br, or I atoms, especially F, Cl, and Br.
  • The phrase "alkyl" refers to groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The phrase "C1-6 alkyl" has the same meaning as alkyl, except that it is limited to alkyl groups of six carbons or less. The phrase C1-6 alkyl. also includes branched chain isomers of straight chain alkyl. groups, including but not limited to, the following which are provided by way of example: -CH(CH3)2, -CH(CH3)(CH2CH3), -CH(CH2CH3)2, -C(CH3)3, -CH2CH(CH3)2, -CH2CH(CH3)(CH2CH3), -CH2CH(CH2CH3)2, -CH2C(CH3)3, -CH(CH3)CH(CH3)(CH2CH3), -CH2CH2CH(CH3)2, -CH2CH2CH(CH3)(CH2CH3), -CH2CH2C(CH3)3, -CH(CH3)CH2CH(CH3)2, -CH(CH3)CH(CH3)CH(CH3), and others. The phrase C1-6 alkyl further includes cyclic alkyl or C3-6 cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and such rings substituted with straight and branched chain alkyl groups as defined above. The phrase alkyl also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • The phrase "aryl" refers to groups that do not contain heteroatoms. The phrase "C6-10 aryl" has the same meaning as aryl, except that it is limited to aryl groups of six to ten carbons atoms. The phrase aryl includes, but is not limited to, groups such as phenyl, biphenyl, and naphthyl by way of example. Aryl groups also include those in which one of the aromatic carbons is bonded to an alkyl, alkenyl, or alkynyl group as defined herein. This includes bonding arrangements in which two carbon atoms of an aryl group are bonded to two atoms of an alkyl, alkenyl, or alkynyl group to define a fused ring system (e.g. dihydronaphthyl or tetrahydronaphthyl). Thus, the phrase "aryl" includes, but is not limited to tolyl, and hydroxyphenyl among others.
  • The phrase "alkenyl" refers to straight chain, branched chain, and cyclic groups such as those described with respect to alkyl groups as defined above, except that at least one double bond exists between two carbon atoms. The phrase "C2-6 alkenyl" has the same meaning as alkenyl, except that it is limited to alkenyl groups of two to six carbons. Examples include, but are not limited to, vinyl, -CH=C(H)(CH3), -CH=C(CH3)2, -C(CH3)=C(H)2, -C(CH3)=C(H)(CH3), -C(CH2CH3)=CH2, cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, hexadienyl, and the like.
  • The phrase "alkoxy" refers to groups having the formula -O-alkyl, wherein the point of attachment is the oxy group and the alkyl group is as defined above. The phrase "C1-6 alkoxy" has the same meaning as alkoxy, except that it is limited to alkoxy groups having from one to six carbon atoms.
  • The phrase "aryloxy" refers to groups having the formula -O-aryl, wherein the point of attachment is the oxy group and the aryl group is as defined above. The phrase "C6-10 aryloxy" has the same meaning as aryloxy, except that it is limited to aryloxy groups of six to ten carbon atoms.
  • The phrase "C1-6 alkoxy-C1-6 alkyl" refers to ether groups with as many as 12 carbon atoms. One example of a C1-6 alkoxy-C1-6 alkyl group is -CH2-O-CH2CH3.
  • The phrase "C6-10 aryloxy-C1-6 alkyl" refers to aryl ether groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the C1-6 alkyl group. One example of a C6-10 aryloxy-C1-6 alkyl group is propoxybenzene.
  • The phrase "C6-10 aryl-C1-6 alkyl" refers to arylalkyl groups of 16 carbon atoms or less, especially of 10 carbon atoms or less bound at the C1-6 alkyl group. One example of a C6-10 aryl-C1-6 alkyl group is toluene.
  • The phrase "amino" refers to NH2.
  • The phrase "alkynyl" refers to straight and branched chain groups such as those described with respect to alkyl groups as defined above, except that at least one triple bond exists between two carbon atoms. The phrase "C2-6 alkynyl" has the same meaning as alkynyl, except that it is limited to alkynyl groups of two to six carbons. Examples include, but are not limited to, -C≡C(H), -C≡C(CH3), -C≡C(CH2CH3), -C(H2)C≡C(H), -C(H)2C≡C(CH3), -C(H)2C≡C(CH2CH3), and the like.
  • The phrase "trihalomethyl" refers to a methyl group in which the three H atoms of the methyl group are substituted with three halogens which may be same or different. One example of such a group is a -CF3 group in which all three H atoms of the methyl group are substituted with F atoms.
  • For clarification, -CH2C(CH3)2(OH) refers to 2-methylpropan-2-ol or tertbutanol.
  • The phrase "heterocyclyl" refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S. Examples of heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g. 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl etc.), tetrazolyl, (e.g. 1H-tetrazolyl, 2H tetrazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing 1 to 4 nitrogen atoms such as, but not limited to, indolyl, isoindolyl, indolinyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms such as, but not limited to furanyl; unsaturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, oxazolyl, isoxazolyl, oxadiazolyl (e.g. 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms such as, but not limited to, morpholinyl; unsaturated condensed heterocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, benzoxazolyl, benzoxadiazolyl, benzoxazinyl (e.g. 2H-1,4-benzoxazinyl etc.); unsaturated 3 to 8 membered rings containing 1 to 3 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolyl, isothiazolyl, thiadiazolyl (e.g. 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, thiazolodinyl; saturated and unsaturated 3 to 8 membered rings containing 1 to 2 sulfur atoms such as, but not limited to, thienyl, dihydrodithiinyl, dihydrodithionyl, tetrahydrothiophene, tetrahydrothiopyran; unsaturated condensed heterocyclic rings containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms such as, but not limited to, benzothiazolyl, benzothiadiazolyl, benzothiazinyl (e.g. 2H-1,4-benzothiazinyl, etc.), dihydrobenzothiazinyl (e.g. 2H-3,4-dihydrobenzothiazinyl, etc.), unsaturated condensed heterocyclic rings containing 1 to 2 oxygen atoms such as benzodioxolyl (e.g. 1,3-benzodioxoyl, etc.); unsaturated 3 to 8 membered rings containing an oxygen atom and 1 to 2 sulfur atoms such as, but not limited to, dihydrooxathiinyl; saturated 3 to 8 membered rings containing 1 to 2 oxygen atoms and 1 to 2 sulfur atoms such as 1,4-oxathiane; unsaturated condensed rings containing 1 to 2 sulfur atoms such as benzothienyl, benzodithiinyl; and unsaturated condensed heterocyclic rings containing an oxygen atom and 1 to 2 oxygen atoms such as benzoxathiinyl. Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones). For example, heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1-dioxide. Preferred heterocyclyl groups contain 5 or 6 ring members. More preferred heterocyclyl groups include morpholine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran. "Heterocyclyl" also refers to those groups as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples, include, but are not limited to, 2-methylbenzimidazolyl, 5-methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1-methyl piperazinyl, and 2-chloropyridyl among others. Heterocyclyl groups are those limited to having 2 to 15 carbon atoms and as many as 6 additional heteroatoms as described above. More preferred heterocyclyl groups have from 3 to 5 carbon atoms and as many as 2 heteroatoms. Most preferred heterocyclyl groups include piperidinyl, pyrrolidinyl, azetidinyl, and aziridinyl groups.
  • The term "substituted" refers to the replacement of one or more hydrogen atom with a monovalent or divalent radical. Suitable substitution groups include, for example, hydroxy, nitro, amino, imino, cyano, halo, thio, thioamido, amidino, imidino, oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamido, carboxyl, formyl, alkyl, heterocyclyl, aryl, haloalkyl, alkoxy, alkoxyalkyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkylthio, aminoalkyl, alkylamino, cyanoalkyl, and the like. For example, one preferred "substituted C1-6 alkyl" is tertbutanol. Other preferred substituted C1-6 alkyl groups include alkoxyalkyl groups (i.e., groups of formula -alkyl-O-alkyl), and - CH2C(CH3)2NH-SO2CH3.
  • The substitution group can itself be substituted one time. For example, an alkoxy substituent of an alkyl group may be substituted with a halogen, and oxo group, an aryl group, or the like. The group substituted onto the substitution group can be carboxyl, halo, nitro, oxo, amino, cyano, hydroxy, C1-6 alkyl, C1-6 alkoxy, C6-10 aryl, aminocarbonyl, -SR, thioamido, -SO3H, -SO2R or cycloalkyl, where R is typically hydrogen, hydroxy or C1-6 alkyl.
  • When the substituted substituent includes a straight chain group, the substitution can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and the like) or at the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like). Substituted substituents can be straight chain, branched or cyclic arrangements of covalently bonded carbon atoms or heteroatoms.
  • The term "protected" or a "protecting group" with respect to hydroxy groups, aminc groups, and sulfhydryl groups refers to forms of these functionalities which are protected from undesirable reaction with a protecting group known to those skilled in the art such as those set forth in Protective Groups in Organic Synthesis, Greene, T.W., John Wiley & Sons, New York, NY, (1st Edition, 1981) which can be added or removed using the procedures set forth therein. Examples of protected hydroxy groups include, but are not limited to, silyl ethers such as those obtained by reaction of a hydroxy group with a reagent such as, but not limited to, t-butyldimethyl-chlorosilane, trimethylchlorosilane, triisopropylchlorosilane, triethylchlorosilane; substituted methyl and ethyl ethers such as, but not limited to methoxymethyl ether, methythiomethyl ether, benzyloxymethyl ether, t-butoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyranyl ethers, 1-ethoxyethyl ether, allyl ether, benzyl ether; esters such as, but not limited to, benzoylformate, formate, acetate, trichloroacetate, and trifluoracetate. Examples of protected amine groups include, but are not limited to, benzyl or dibenzyl, amides such as, formamide, acetamide, trifluoroacetamide, and benzamide; imides, such as phthalimide, and dithiosuccinimide; and others. In some embodiments, a protecting group for amines is a benzyl group. Examples of protected sulfhydryl groups include, but are not limited to, thioethers such as S-benzyl thioether, and S-4-picolyl thioether; substituted S-methyl derivatives such as hemithio, dithio and aminothio acetals; and others.
  • Imidazoquinoxaline compounds of Formula (I) or (II) and the analogs thereof may exhibit the phenomenon of tautomerism, and the formula drawings within this specification can represent only one of the possible tautomeric forms. It is to be understood that the invention encompasses any tautomeric form which possesses immunomodulatory activity and is not to be limited merely to any one tautomeric form utilized within the formula drawings.
  • Imidazoquinoxalines of Formula (I) or (II) also may exist in solvated as well as unsolvated forms such as, for example, hydrated forms. The invention encompasses both solvated and unsolvated forms which possess immunomodulatory activity.
  • The invention also includes isotopically-labeled imidazoquinoxaline compounds and analogs thereof, that are structurally identical to those disclosed above, except that one or more atom is/are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 18O 17O, 31P, 32P, 35S 18F and 36Cl, respectively. Compounds of the present invention, tautomers thereof, prodrugs thereof, and pharmaceutically acceptable salts of the compounds and of the prodrugs that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes such as3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out known or referenced procedures and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • The foregoing may be better understood by reference to the following Examples that are presented for illustration and not to limit the scope of the inventive concepts. The Example compounds and their analogs are easily synthesized by one skilled in the art from procedures described herein, as well as in patents or patent applications listed herein which are all hereby incorporated by reference in their entireties and for all purposes as if fully set forth herein.
  • EXAMPLES
  • Compounds of the embodiments may generally be prepared using a number of methods familiar to one of skill in the art, such as, for example, the methods disclosed in the literature publications "Imidazo[1,2-a]quinoxalines: synthesis and cyclic nucleotide phosphodiesterase inhibitory activity." Stephanie Parra, et. al. European Journal of Medicinal Chemistry, 2001, 36, pp. 255-264; "Design, synthesis and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine Al receptor antagonists." Chun-He Liu, et. al. Bioorganic and Medicinal Chemistry, 2004, 12, pp. 4701-4707; "Design and synthesis of novel imidazo[1,2-a]quinoxaolines as PDE4 inhibitors." Carine Deleuze-Masquefa, et. al. Bioorganic and Medicinal Chemistry, 2004, 12, pp. 1129-1139; and references therein. The disclosures of each of the foregoing are incorporated herein in their entirety for all purposes. The compounds of the embodiments may be generally made in accordance with the following reaction Schemes 1-2, which are described in detail below.
  • Example 1 General Preparative Routes for the Synthesis of Selected Compounds of the Invention
  • Schemes 1-2 illustrate general methods for the preparation of intermediates and compounds of the embodiments. These compounds are prepared from starting materials either known in the art for commercially available. The specific compounds are for illustrative purposes only.
    Figure imgb0033
  • As seen in Scheme 1, 2,3-dichloropyrazines (1.1), or equivalent reagents, reacts with a substituted or unsubstituted oc-hydroxyalkylamine (1.2) to give a intermediates of Formula 1.3. Oxidation of the alcohol of intermediates 1.3 under conditions known to those familiar in the art as a Swern oxidation, or equivalents, provides the aldehyde or ketone intermediates of Formula 1.4. Treatment with a dehydrating reagent such as, for example, TFAA in a solvent such as, for example, TFA then provides the intermediates of Formula 1.5. Treatment of intermediate 1.5 with ammonia then provides compounds of Formula 1.6. Alternately, intermediate 1.5 can be treated with reagents such as hydrazine, hydroxyamine, sodium azide or equivalents to provide intermediates of Formula 1.7. Treatment with under reducing conditions such as, for example, hydrogen with palladium on carbon in a solvent such as methanol or tetrahydrofuran then provides compounds of Formula 1.6. Alternately, intermediate 1.5 can be treated with a suitably protected amine to provide intermediates of Formula 1.8. Removal of the protecting groups provides compounds of the Formula 1.6.
  • The 2.3-dichlorpyrazines of Formula 1.1 can be obtained commercially or by one trained in the art following chemistries similar to those found in "Product class 14: pyrazines." N. Sato. Science of Synthesis 2004, 16, pp. 751-844; "Pyrazines. I. Syntheses of 2,3-dihydroxypyrazines and their derivatives." Jiro Adachi, Nobuhiro Sato. Journal of Organic Chemistry 1972, 37(2), pp. 221-5; "Displacements and nuclear substitutions on hydroxypyrazines." Geo Karmas. Paul E, Spoerri. Journal of the American Chemical Society 1956, 78, pp. 4071-7; each of which is incorporated by referene herein in its entiety for all purposes.
    Figure imgb0034
  • Scheme 2 depicts alternate methods for the preparation of compounds of the embodiment through an intermediate of Formula 2.1 prepared by methods described in Scheme 1. Treatment of intermediates of the Formula 2.1 with a halogenating agent such as, for example, NBS provides the halogenated intermediate 2.2 which can be treated with methods, known to one trained in the art, for the removal of protecting groups to provide compounds of the embodiment of Formula 2.3. Other halogenating agents can be used to in the conversion of intermediates of Formula 2.1 to 2.2. Other halogenating agents include, but are not limited to, NIS, NCS, chlorine, bromine, iodine and ICl.
  • Deprotection of a compound of Formula 2.2 where P and P' are 4-methoxybenzyl groups treatment with an acid such as, for example, HCl in a solvent such as, for example, dioxane or THF can provide compounds of the embodiment of Formula 2.3. In another example an intermediate of Formula 2.2 where P and P' are taken together to form a phthalamide, or equivalent protecting group, can be treated with a reagent such as, for example, hydrazine in a solvent such as, for example, methanol, tetrahydrofuran or dimethylformamide can provide compounds of the embodiment of Formula 2.3.
  • Treatment of compounds of Formula 2.1 with a strong base such as, for example, butyllithium in a solvent such as tetrahydrofuran followed by introduction of an electrophile such as, for example, methyl iodide or propylene oxide provides intermediates of the Formula 2.4. Additionally, intermediates of Formula 2.2 can be treated with a reagent such as, for example, tert-butyllithium in a solvent such as, for example, tetrahydrofuran followed by addition of an electrophile such as, for example, methyl iodide or propylene oxide provides intermediates of the Formula 2.4. Furthermore, the halo functionality of intermediate 2.2 in conjunction with transition metal mediated couplings such as those described in "Palladium Reagents and Catalysts: Innovations in Organic Chemistry" by Jiro Tsuji. John Wiley & Sons Ltd., West Susses, England, 1995; "Oranopalladium Chemistry for Organic Synthesis" by E. Negishi. John Wiley & Sons Ltd., New York, NY, 2002; and references therein, provides access to intermediates of Formula 2.4. Deprotection of P and P' as previously described then provides compounds of the embodiment of Formula 2.5.
  • Example 2 General Preparative Routes for the Synthesis of 1-(4-amino-2-isobutylimidazo[1,2-a]quinoxalin-1-yl)-2-methylpropan-2-ol and 2-isobutylimidazo[1,2-a]quinoxalin-4-amine
  • Figure imgb0035
    Figure imgb0036
  • Commercially available 2,3-dichloropyrazine (3.1), is reacted with reacts with the α-hydroxyalkylamine (3.2) to give the intermediate of Formula 3.3. Oxidation of the alcohol of intermediates 3.3 with a reagent such as, for example, SO3.pyridine in a solvent such as, for example, DMSO provides the aldehyde intermediate of Formula 3.4. Treatment with a dehydrating reagent such as, for example, TFAA in a solvent such as, for example, TFA then provides the intermediates of Formula 3.5. Intermeidates of the Formula 3.5 can be treated with a suitably protected amine such as, for example, bis(4-methoxybenzyl)amine to provide intermediates of Formula 3.6. Removal of the 4-methoxybenzyl protecting groups with a reagents such as, for example, TFA provides compounds of the embodiment of Formula 3.7. In addition, intermediates of the formual 3.6 can be treated with a strong base such as, for example, butyllithium in a solvent such as, for example, tetrahydrofuran followed by the addition of an electrophile such as, for example, isopropylene oxide to provide intermediates of the formua 3.8. Removal of the 4-methoxybenzyl protecting groups with a reagents such as, for example, TFA provides compounds of the embodiment of Formula 3.9. The detailed preparaiton of these compounds is shown in Examples 3 and 4, below.
  • Example 3 Synthesis of 2-isobutylimidazo[1,2-a]quinoxalin-4-amine
  • The title compound was synthesized according to Scheme 4 below:
    Figure imgb0037
  • A. Synthesis of 2-(3-chloroquinoxalin-2-ylamino)-4-methylpentan-1-ol
  • Figure imgb0038
  • The title compound was synthesized generally according to the procedures described in Carine Deleuze-Masquefa, Grégori Gerebtzoff, Guy Subra, Jean-Roch Fabreguettes , Annabel Ovens, Maëlle Carraz, Marie-Paule Strub, Jacques Bompart. Design and synthesis of novel imidazo[1,2-a]quinoxalines as PDE4 inhibitor. Bioorganic & Medicinal Chemistry (2004), 12(5), 1129-1139. and in Chun-He Liu, Bo Wang, Wei-Zhang Li, Liu-Hong Yun, Ying Liu, Rui-Bing Su, Jin Li and He Liu. Design, synthesis, and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine A1 receptor antagonist. Bioorganic & Medicinal Chemistry (2004), 12(17), 4701-4707.
    To a solution of 2,3-dichloroquinoxaline (2.0 g, 10.0 mmol) in 10 mL of 1,4-dioxane was added triethyl amine (1.81 g, 18 mmol) and followed by 2-amino-4-methylpentan-1-ol ( 1.4 g, 12 mmol). The reaction mixture was separated into 2 microwave tubes. And then the mixture was stirred in microwave for 10 minutes at 130°C. The combined reaction mixture was diluted with 150 mL of ethyl acetate and then was washed with water (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography eluted with ethyl acetate and hexanes to give the titled compound (2.3g, 69%)
    MS: MH+ = 280 : 282 = 3 : 1
  • B. Synthesis of 2-(3-chloroquinoxalin-2-ylamino)-4-methylpentanal
  • Figure imgb0039
  • The title compound was synthesized generally according to the procedures described in Chun-He Liu, Bo Wang, Wei-Zhang Li, Liu-Hong Yun, Ying Liu, Rui-Bing Su, Jin Li and He Liu. Design, synthesis, and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine A1 receptor antagonist. Bioorganic & Medicinal Chemistry (2004), 12(17), 4701-4707.
  • To a solution of the 2-(3-chloroquinoxalin-2-ylamino)-4-methylpentan-1-ol (1.90 g, 6.79 mmol) in 10 mL of DMSO was added triethyl amine (1.92 g, 19.0 mmol) and followed by sulfur trioxide pyridine complex (2.38 g, 14.9 mmol) at room temperature. The reaction mixture was stirred at that temperature overnight. And then 100 mL of water was added to reaction mixture. The resulting mixture was extracted with ethyl acetate (75 mL x 2). The combined organic layers were washed with water (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography eluted with ethyl acetate and hexanes to give the titled compound (1.25 g, 64%). MS: MH+ = 278 : 280 = 3 : 1
  • C. Synthesis of 4-chloro-2-isobutylimidazo[1,2-a]quinoxaline
  • Figure imgb0040
  • To a solution of the 2-(3-chloroquinoxalin-2-ylamino)-4-methylpentanal (1.03 g, 3.71 mmol) in 8 mL of trifluoroacetic anhydride was added 0.2 mL trifluoroacetic acid at room temperature. The reaction mixture was stirred at that temperature overnight. The solvents were removed under reduced pressure. And then 20 mL of saturated aqueous sodium bicarbonate was added. The resulting mixture was extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with water (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography eluted with ethyl acetate and hexanes to give the titled compound (630 mg, 65%). MS: MH+ = 260 : 262 = 3 : 1. 1H NMR (300 MHz, CDC13): δ 8.05 (m, 1H), 7.93 (s, 1H), 7.86 (m, 1H), 7.67 (m, 1H), 7.59 (m, 1H), 2.87 (d, 2H), 2.17 (m, 1H), 1.02 (d, 6H).
  • D. Synthesis of 2-isobutyl-N,N-bis(4-methoxybenzyl)imidazo[1,2-a]quinoxalin-4-amine
  • Figure imgb0041
  • To a solution of the 4-chloro-2-isobutylimidazo[1,2-a]quinoxaline (130 mg, 0.50 mmol) in 2 mL of 1-methylpyrrolidin-2-one was added N,N-diisopropylethylamine (129 mg, 1.0 mmol) and followed by bis(4-methoxybenzyl)amine (154 mg, 0.60 mmol). The reaction mixture was stirred at 120°C overnight. And then 10 mL of water was added. The resulting mixture was extracted with ethyl ether (50 mL × 3). The combined organic layers were washed with water (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography eluted with ethyl acetate and hexanes to give the titled compound (195 mg, 81 %). MS: MH+ = 481. 1H NMR (300 MHz, CDC13): δ 7.72 (s, 1H), 7.65 (m, 2H), 7.21-7.39 (m, 6H), 6.84 (d, 4H), 5.34 (brs, 4H), 3.79 (s, 6H), 2.63 (d, 2H), 2.06 (m, 1H), 0.94 (d, 6H).
  • E. Synthesis of 2-isobutylimidazo[1,2-a]quinoxalin-4-amine
  • Figure imgb0042
  • The solution of 2-isobutyl-N,N-bis(4-methoxybenzyl)imidazo[1,2-a]quinoxalin-4-amine (16 mg, 0.033 mmol) in 1 mL of trifluoroacetic acid was stirred at 70°C for 6 hours. The solvent was removed under reduced pressure. And then 10 mL of saturated aqueous sodium bicarbonate was added. The resulting mixture was extracted with 75 mL of ethyl acetate. The organic layer was separated and then washed with water (15 mL), brine (15 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by preparative TLC sheet (ethyl acetate and hexanes) to give the titled compound (6.3 mg, 79%). MS: MH+ = 241. 1H NMR (300 MHz, CDCl3): δ 7.74 (s, 1H), 7.68 (m, 2H), 7.30-7.40 (m, 4H), 5.70 (brs, 2H), 2.67 (d, 2H), 2.08 (m, 1H), 1.00 (d, 6H).
  • Example 4 Synthesis of 1-(4-amino-2-isobutylimidazo[1,2-a]quinoxalin-1-yl)-2-methylpropan-2-ol A. Synthesis of 1-(4-(bis(4-methoxybenzyl)amino)-2-isobutylimidazo[1,2-a]quinoxalin-1-yl)-2-methylpropan-2-ol
  • Figure imgb0043
  • To a solution of the 2-isobutyl-N,N-bis(4-methoxybenzyl)imidazo[1,2-a]quinoxalin-4-amine (134 mg, 0.28 mmol) (Example 1, D) in 2 mL of tetrahydrofuran was added n-butyl lithium (2.5M, 0.2 mL, 0.50 mmol) at -78°C. After being stirred at that temperature for 1 hour, 1,2-epoxy-2-methylpropane (72 mg, 1.0 mmol) was added. The reaction mixture was stirred at -78°C for 2 hours, and then at room temperature for 4 hours. The reaction was quenched with 10 mL of saturated aqueous ammonium chloride. The resulting mixture was extracted with 100 mL of ethyl acetate. The organic layer was separated and washed with water (20 mL), brine (20 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography eluted with ethyl acetate and hexanes to give the titled compound (35 mg, 19%) and recovered starting material (100 mg, 63%). MS: MH+ = 553. 1H NMR (300 MHz, CDCl3): δ 8.41 (dd, 1H), 7.67 (dd, 1H), 7.20-7.37 (m, 6H), 6.84 (m, 4H), 5.31 (brs, 4H), 3.79 (s, 6H), 3.51 (s, 2H), 2.58 (d, 2H), 2.14 (m, 1H), 1.37 (s, 6H), 0.88 (d, 6H).
  • B. Synthesis of 1-(4-amino-2-isobutylimidazo[1,2-a]quinoxalin-1-yl)-2-methylpropan-2-ol
  • Figure imgb0044
  • The solution of 1-(4-(bis(4-methoxybenzyl)amino)-2-isobutylimidazo[1,2-a]quinoxalin-1-yl)-2-methylpropan-2-ol (15 mg, 0.027 mmol) in 1 mL of trifluoroacetic acid was stirred at 70°C for 6 hours. The solvent was removed under reduced pressure. And then 10 mL of saturated aqueous sodium bicarbonate was added. The resulting mixture was extracted with 70 mL of ethyl acetate. The organic layer was separated and then washed with water (15 mL), brine (15 mL), drie over MgSO4, filtered and concentrated. The crude product was purified by preparative TLC sheet (ethyl acetate and hexanes) to give the titled compound (6.5 mg, 77%). MS: MH+ = 313. 1H NMR (300 MHz, CDC13): δ 8.41 (dd, 1H), 7.64 (dd, 1H), 7.25-7.41 (m, 2H), 5.63 (brs, 2H), 3.51 (s, 2H) 2.64 (d, 2H), 2.21 (m, 1H), 1.37 (s, 6H), 0.95 (d, 6H).
  • Each of the following compounds are readily synthesized according to the above Examples
    Figure imgb0045
    Figure imgb0046
    Figure imgb0047
    Figure imgb0048
    Figure imgb0049
    Figure imgb0050
    Figure imgb0051
  • The compounds of Examples 3 and 4 exhibited an EC50 value with respect to agonism of IL-6 and TNF-α at less than 15 µM. The compounds of Examples 3 and 4 were tested for stimulatory activity of human TLR 7 and 8 receptors and the mouse TLR7 receptor. Both compounds stimulated TLR7 and TLR8 activity, wherein the compound of Example 3 exhibited slightly more reactivity for mouse TLR7 than did the compound of Example 4. The compound of Example 3 exhibited stimulatory activity with human TLR8 while the compound fo Example 4 exhibited little or no reactivity with human TLRB. The compound of Example 3, and to a greater extent, Example 4 exhibited activity for stimulation of human TLR7. Additionally, because of the excellent activity of the compounds, each of these compounds is individually preferred and is preferred as a member of a group that includes any or all of the other compounds of Formula (I), or (II), or both. Each compound is preferred in methods of modulating an immune response and in methods of treating biological conditions associated therewith, for example to be used as a vaccine adjuvant. Each of the compounds is also preferred for use in preparation of medicaments for immunopotentiation, reducing tumor growth, treating microbial and viral infections, particularly influenza, HCV and HSV, and in treating biological conditions mediated therefrom.
  • Compounds found to not be effective at a concentration of 20 µM or less using the assay described below are also useful within the scope of the invention, since the invention is not meant to be limited to those compounds that are useful at a concentration of 20µM or less. Compounds may be useful as intermediates, or as final products that cause production of TNF-α at higher concentrations, such as 100 µM, 200 µM or 300 µM in the assays described herein. For example Loxoribine causes useful production of TNF-α at 300 µM (see Pope et al. Cellular Immunology 162: 333-339 (1995)).
  • BIOLOGICAL ASSAYS
  • Candidate small molecule immunopotentiators can be identified in vitro. Compounds are screened in vitro for their ability to activate immune cells. One marker of such activation is the induction of cytokine production, for example TNF-α production. Apoptosis inducing small molecules may be identified having this activity. These small molecule immuno-potentiators have potential utility as adjuvants and immuno-therapeutics.
  • In an assay procedure (High Throughput Screening (HTS)) for imidazoquinoxaline small molecule immune potentiators (SMIPs), human peripheral blood mononuclear cells (PBMC), 500,000 per mL in RPMI 1640 medium with 10% FCS, are distributed in 96 well plates (100,000 per well) already containing 5 µM of compound in DMSO. The PBMCs are incubated for 18 hours at 37°C in 5% CO2. Their ability to produce cytokines in response to the small molecule compounds is determined using a modified sandwich ELISA.
  • Briefly, supernatants from the PBMC cultures are assayed for secreted TNF using a primary plate bound antibody for capture followed by a secondary biotinylated anti-TNF antibody forming a sandwich. The biotinylated second antibody is then detected using streptavidin-europium, and the amount of bound europium is determined by time resolved fluorescence. Imidazoquinoxaline compounds and analogs thereof are confirmed by their TNF inducing activity that is measured in the assay as increased europium counts over cells incubated in RPMI medium alone. "Hits" are selected based on their TNF-inducing activity relative to an optimal dose of lipopolysaccharide LPS (1 µg/mL), a strong TNF inducer. The robustness of the assay and low backgrounds allow for the routine selection of hits with ~10% of LPS activity that is normally between 5-10X background (cells alone). Selected hits are then subjected to confirmation for their ability to induce cytokines from multiple donors at decreasing concentrations. Those compounds with consistent activity at or below 5 µM are considered confirmed for the purposes of this assay. The assay is readily modified for screening for compounds effective at higher or lower concentrations.
  • In addition to the procedure described above, methods of measuring other cytokines (e.g., ILl-beta, IL-12, IL-6, IFN-gamma, IL-10 etc.) are well known in the art and can be used to find active imidazoquinoxaline compounds and analogs thereof of the present invention.
  • Qualitative and quantitative measurement of the immune response of a SMIP or composition comprising a SMIP of the preferred embodiments of the present invention can be implemented using methods known in the art, such as by measuring antigen specific antibody production, activation of specific populations of lymphocytes such as CD4+ , CD8+ T cells or NK cells, and/or production of cytokines such as IFN, IL-2, IL-4 or IL-12. Methods for measuring specific antibody responses include enzyme-linked immunosorbent assay (ELISA) as known in the art. Measurement of numbers of specific types of lymphocytes such as CD4+ T cells can be achieved, for example, with fluorescence-activated cell sorting (FACS). Cytotoxicity assays can also be performed using methods known in the art, e.g., as described in Raz et al., (1994) Proc. Natl. Acad. Sci. USA 91:9519-9523. Serum concentrations of cytokines can be measured, for example, by ELISA. Such assays are described, e.g., in Selected Methods in Cellular Immunology (1980) Mishell and Shiigi, eds., W.H. Freeman and Co.
  • I. Sample Preparations Human PBMC preparation
  • Human blood from one or multiple human donors were collected into the BD Vacutainer™ CPT tube with sodium citrate (BD, Franklin Lakes, NJ), and spun for 20 minutes at 1600g. After centrifugation, mononuclear cells in the top layer in the tubes were collected and then washed three times with PBS buffer. The washed cells were then reconstituted at a required cell concentration in complete RPMI containing 10% FBS plus 100 units/ml penicillin and 100ug/ml streptomycin.
  • Mouse Spleen Cell Preparation
  • Spleens were isolated from Balbc mice and minced to release the splenocytes from the tissues. After the minced samples were treated ammonium salt to destroy the red blood cells, the rest of the spleenocytes were washed and reconstituted at a required cell concentration with completed RPMI medium.
  • Human THP-1 Cell Line
  • The human myelomonocytic transformed cell line is responsive to TLR8 agonists and weakly to TLR7 agonists. The cell line is cultured in RPMI medium supplemented with 10% FBS.
  • II. Activity Measurement Compound Stimulation and Multi-cytokine Measurement
  • Human PBMC (hPBMC) (at 1 million cells/ml) or mouse spleen cells (at 5 million cells/ml) or human monocytic THP-1 cells (at 1 million cells/ml) were mixed with tested compounds such as imidazoquinoxalines at titrated compound concentrations in the complete RPMI medium. After the cell cultures were incubated for 24 hours at 37°C, 5% CO2, the culture supernatant was collected and assayed for the secreted cytokines in the presence of the compounds. Human or mouse Beadlyte multi-cytokine flex kits (Upstate, Lake Placid, NY) were used to measure the amount of the following cytokines: TNF-α and IL-6 according to the manufacturers instructions.
  • TLR Signaling
  • HEK293 cells (ATCC, CRL-1573) are seeded in a T75 flask at 3x106 in 20ml of DMEM supplemented with 0.1mM nonessential amino acid, 1mM sodium pyruvate, 2mM L-glutamine, penicillin-streptomycin, and 10% FCS. After overnight culturing, the cells are transfected with 1) pNFkB-TA-luciferase reporter (0.4ug) (BD clontech, Palo Alto, CA), and with 2) with pGL4.74 (0.01ug) that carries a TK promoter, not responsive to NF-kB stimulation, and carries a Renilla luciferase gene, used as an internal control (Promega, WI), and 3), separately with a following TLR construct (10 ug): human TLR (hTLR) 7, hTLR8, mouse TLR7 (mTLR7) puno constructs (Invivogene, CA), using Fugene 6 transfection reagent (Roche). The transfected cells after 24 hours transfection are collected and seeded in a 96-well and flat-bottom plate (1x104 cell/well) plate, and stimulated with the test compounds at the following concentrations: 30, 10, 3, 1, 0.3, 0.1, 0.03 uM. After overnight compound stimulation, the cells are assayed for expression of fly and renilla luciferases using Dual-Luciferase Reporter Assay System (Promega, WI). NF-kb activation is directly proportional to relative fly luciferase units, which is measured against the internal control renilla luciferase units.
  • Standardization of Cytokine Production
  • Due to the agonist nature of the compounds tested, compound ranking is based on potency in cell-based screens for cytokine induction. Briefly, the EC50 of each compound for a given cytokine is calculated relative to a reference composition (i.e. LPS or other potent immune stimulator). This value is then used as the divisor of the maximum level of cytokine produced (pg/ml) in the assay. Five parameter curve fitting of cytokine dose response curves to different SMIPs for the indicated cell populations is used to calculate EC50. Rank-scoring of SMIP potency is calculated by dividing the maximum concentration of cytokine produced by the relative EC50 established for each compound indicated.
  • In Vivo Adjuvant Studies
  • In phosphate-buffered saline (PBS), 25 micrograms gp120dV2EnvSF162 antigen (recombinant gp120 protein derived from sequence of HIV-1 strain SF162 - the V2 domain is deleted; Pharm Res. 2004 Dec 21(12):2148-52) as mixed with 50 microliters of MF59 adjuvant, followed the by the addition of 0, 1, 5, or 25 micrograms of a small molecule immune potentiator (SMIP) and adjusted to 100 microliters with PBS. 50 microliters of the solution is subsequently injected into both the left and right tibialis anterior muscles of female BALB/c mice (Day 0), for a total volume of 100 microliters per mouse. Four weeks later (Day 28), 50 microliters of the solution is again injected into both the left and right tibialis anterior muscles of the mouse. Seven days after the second vaccination (Day 34), serum samples ae collected, and a day later (day 35) spleens are removed. Serum samples are assayed by Env-specific serum IgG2a ELISA and Env-specific serum IgG1 ELISA. Spleen samples are assayed by Env-specific, cytokine-producing splenic CD4 and CD8 T cells.
  • The contents of each of the patents, patent applications and journal articles cited above are hereby incorporated by reference herein and for all purposes as if fully set forth in their entireties.
  • Aspects of the invention
    1. 1. A compound of Formula (1):
      Figure imgb0052
      wherein:
      • R1 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, -NR3C(=O)NR4R5, - NR3S(=O)pNR4Rs, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
      • R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4, - NR3C(=O)NR4Rs, -NR3S(=O)pNR4Rs, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
      • R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl;
      • R6 is selected from the group consisting of hydrogen, halogen, hydroxy, -OR3, - N(R3)(R4), C1-3 haloalkyl, -O-C1-3 haloalkyl, C1-3 perhaloalkyl, -O-C1-3 perhaloalkyl, C1-3 hydroxyalkyl, -O-C1-3 hydroxyalkyl, CN, -(CH2)qC(=O)R7, -O-(CH2)qC(=O)R7, - (CH2)qN(R8)(R9), -S-C1-3alkyl, -S(=O)2-R10 and -S(=O)2N(R8)(R9);
      • q is 0,1, 2 or 3;
      • R7 is selected from hydrogen, hydroxy, C1-3 alkyl or C1-3 alkoxy; and
      • R8, R9 and R10 are each independently hydrogen or C1-3 alkyl;
      • provided that R1, R2 and R6 are not simultaneously hydrogen; and
      • if R1 and R6 are both H, R2 is not butyl;
      or a pharmaceutically acceptable salt thereof; a tautomer thereof, or a pharmaceutically acceptable salt of the tautomer.
    2. 2. A compound of aspect 1, wherein R1 is -N(R3)(R4), C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, or substituted C2-C12 alkenyl.
    3. 3. A compound of aspect 1, wherein R1 is hydrogen, C1-C12 alkyl or substituted C1-C12 alkyl.
    4. 4. A compound of aspect 1, wherein R1 is C1-C12 alkyl.
    5. 5. A compound of aspect 1, wherein R1 is C1-C6 alkyl.
    6. 6. A compound of aspect 1, wherein R1 is -CH2-CH(CH3)2.
    7. 7. A compound of aspect 1, wherein R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl.
    8. 8. A compound of aspect 2, wherein R2 is C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl.
    9. 9. A compound of aspect 1, wherein R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl.
    10. 10. A compound of aspect 1, wherein R2 is hydroxyalkyl.
    11. 11. A compound of aspect 1, wherein R2 is hydrogen.
    12. 12. A compound of aspect 1, wherein R2 is -CH2-C(OH)(CH3)2.
    13. 13. A compound of aspect 1, wherein:
      • R1 is C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, or substituted C2-C12 alkenyl; and
      • R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl.
    14. 14. A compound of aspect 1, wherein:
      • R1 is C1-C12 alkyl or substituted C1-C12 alkyl; and
      • R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl or hydroxyalkyl.
    15. 15. A compound of aspect 1, wherein:
      • R1 is C1-C12 alkyl; and
      • R2 is hydrogen, C1-C12 alkyl or hydroxyalkyl.
    16. 16. A compound of aspect 1, wherein:
      • R1 is C1-C6 alkyl; and
      • R2 is hydrogen or hydroxyalkyl.
    17. 17. A compound of aspect 1, wherein:
      • R1 is -CH2-CH(CH3)2; and
      • R2 is hydrogen or hydroxyalkyl.
    18. 18. A compound of aspect 1, wherein:
      • R1 is -CH2-CH(CH3)2; and
      • R2 is hydrogen or -CH2-C(OH)(CH3)2.
    19. 19. A compound of any of aspects 1-18, wherein R6 is hydrogen.
    20. 20. A compound of aspect 1, wherein R6 is -OR3 or -N(R3)(R4).
    21. 21. A compound of aspect 1, wherein only one of R1, R2 and R6 is H.
    22. 22. A compound of formula:
      Figure imgb0053
      or a pharmaceutically acceptable salt thereof.
    23. 23. A compound of formula:
      Figure imgb0054
      or a pharmaceutically acceptable salt thereof.
    24. 24. A method of inducing interferon biosynthesis in a subject, comprising: administering to the subject a compound according to any one of aspects 1-23 in an amount sufficient to induce interferon biosynthesis.
    25. 25. A method of modulating an immune response in a subject, comprising: administering a compound according to any one of aspects 1-23.
    26. 26. A method for inducing the production of TNF-α in a subject, comprising: administering a compound according to any one of aspects 1-23 to the subject in an amount sufficient to induce the production of TNF-α in the subject.
    27. 27. The method of aspect 26, wherein the compound has an average steady-state drug concentration in the blood of less than 20 µM.
    28. 28. A method of inducing an immune response in a subject, comprising: administering a compound according to any one of aspects 1-23 to the subject in an amount sufficient to induce an immune response in the subject.
    29. 29. The method of aspect 28, wherein the immune response involves the production of cytokines.
    30. 30. The method of aspect 28, wherein the immune response involves modulation of TLR-7 and/or TLR-8.
    31. 31. The method of aspect 28, wherein the subject is suffering from a microbial infection.
    32. 32. The method of aspect 28, wherein the subject is suffering from a viral infection.
    33. 33. The method of aspect 32, wherein the viral infection is a viral infection caused by the hepatitis C virus (HCV).
    34. 34. The method of aspect 32, wherein the viral infection is caused by the human immunodeficiency virus (HIV).
    35. 35. The method of aspect 28, wherein the subject is suffering from abnormal cellular proliferation or cancer.
    36. 36. The method of aspect 28, wherein the subject is suffering from allergic diseases.
    37. 37. The method of aspect 28, wherein the subject is suffering from asthma.
    38. 38. The method of aspect 28, wherein the subject is suffering from precancerous lesions.
    39. 39. The method according to aspect 38, wherein the precancerous lesions are actinic keratosis.
    40. 40. A method of inhibiting a kinase, comprising: administering the compound according to any one of aspects 1-23 to a subject, wherein the kinase is inhibited in the subject.
    41. 41. The method according to any one of aspects 25, 26, 27, 29-36, 38 or 39 wherein the compound is administered topically.
    42. 42. A pharmaceutical composition, comprising: the compound of any one of aspects 1-23 and a pharmaceutically acceptable excipient.
    43. 43. A method of inducing an immune response in a subject, comprising: administering to the subject a compound according to any one of aspects 1-23 and an antigen, wherein the compound induces an immune response to the antigen in the subject.
    44. 44. A method of enhancing the immune response to an antigen in a subject, comprising: administering to the subject a composition comprising a compound according to any one of aspects 1-23 and an antigen, wherein the immune response to the antigen in the subject is enhanced.
    45. 45. A composition comprising the compound according to any one of aspects 1-23 and an additional immunogenic composition or an antigen.
    46. 46. The composition of aspect 45, wherein the additional immunogenic composition comprises an antigen.
    47. 47. The composition according to one of any one of aspects 42, 45 or 46 further comprising an additional adjuvant.
    48. 48. The composition of aspect 47 wherein the adjuvant is MF59.
    49. 49. The composition according to any one of aspects 45-47, further comprising poly(lactide-co-glycolide) (PLG).
    50. 50. The composition according to aspect 46, wherein the antigen is a bacterial antigen or a viral antigen.
    51. 51. The composition according to aspect 50, wherein the antigen is a viral antigen from a virus selected from the group consisting of Hepatitis C virus, Human Immunodeficiency virus, Hepatitis B virus, Human Papiloma virus and Influenza virus.
    52. 52. The composition according to aspect 51, wherein the antigen is an influenza antigen.
    53. 53. The composition of aspect 52 wherein the influenza antigen comprises haemagglutinin and/or neuraminidase surface proteins.
    54. 54. The composition according to one of any one of aspects 50-53 further comprising an additional adjuvant.
    55. 55. The composition of aspect 54 wherein the adjuvant is MF59.
    56. 56. The composition according to any one of aspects 50-55, further comprising poly(lactide-co-glycolide) (PLG).
    57. 57. A composition comprising the compound according to any one of aspects 1-23 and an antigen.
    58. 58. The composition of aspect 57 further comprising an additional adjuvant.
    59. 59. The composition of aspect 58 wherein the adjuvant is MF59.
    60. 60. The composition according to any one of aspects 57-59, further comprising poly(lactide-co-glycolide) (PLG).
    61. 61. The composition according to aspect 57, wherein the antigen is a bacterial antigen or a viral antigen.
    62. 62. The composition according to aspect 61, wherein the antigen is a viral antigen from a virus selected from the group consisting of Hepatitis C virus, Human Immunodeficiency virus, Hepatitis B virus, Human Papiloma virus and Influenza virus.
    63. 63. The composition according to aspect 57, wherein the antigen is an influenza antigen.
    64. 64. The composition of aspect 63 wherein the influenza antigen comprises haemagglutinin and/or neuraminidase surface proteins.
    65. 65. The composition according to one of any one of aspects 61-64 further comprising an additional adjuvant.
    66. 66. The composition of aspect 65 wherein the adjuvant is MF59.
    67. 67. The composition according to any one of aspects 61-66, further comprising poly(lactide-co-glycolide) (PLG).
    68. 68. An immunogenic composition comprising an antigen and an imidazo[1,2-a]quinoxalin-4-amine effective to stimulate a cell mediated immune response to said antigen.

Claims (14)

  1. A compound of Formula (I):
    Figure imgb0055
    wherein:
    R1 is selected from the group consisting of hydrogen
    R2 is selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo, haloalkyl, amino,-OR3, -N(R3)(R4), -NR3C(=O)R4, - NR3S(=O)pR4,-NR3C(=O)NR4R5, -NR3S(=O)pNR4R5, -C(=O)R4, -S(=O)pR4, -C(=O)NR3R4, -S(=O)pNR3R4 and -C(=O)OR4;
    R3, R4 and R5 are each independently selected from the group consisting of hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C3-C12 cycloalkyl, substituted C3-C12 cycloalkyl, aryl, substituted aryl, heterocyclyl, substituted heterocyclyl, heteroaryl, substituted heteroaryl, hydroxy, hydroxyalkyl, halo and haloalkyl-,
    R6 is selected from the group consisting of hydrogen, halogen, hydroxy, -OR3, - N(R3)(R4), C1-3 haloalkyl, -O-C1-3 haloalkyl, C1-3 perhaloalkyl, -O-C1-3 perhaloalkyl, C1-3 hydroxyalkyl, -O-C1-3 hydroxyalkyl, CN, -(CH2)qC(=O)R7, -O-(CH2)qC(=O)R7, - (CH2)qN(Rs)(R9), -S-C1-3 alkyl, -S(=O)2-R10 and -S(=O)2N(R8)(R9);
    q is 0, 1, 2 or 3;
    R7 is selected from hydrogen, hydroxy, C1-3 alkyl or C1-3 alkoxy; and
    R8, R9 and R10 are each independently hydrogen or C1-3 alkyl;
    provided that R1, R2 and R6 are not simultaneously hydrogen; and
    if R1 and R6 are both H, R2 is not butyl;
    or a pharmaceutically acceptable salt thereof; a tautomer thereof; or a pharmaceutically acceptable salt of the tautomer.
  2. A compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, of claim 1, wherein:
    i) R2 is hydrogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, or hydroxyalkyl; or
    ii) R2 is C1-C12 alkyl, substituted C1-C12 alkyl, or hydroxyalkyl; or
    iii) R2 is hydrogen, C1-C12 alkyl, or hydroxyalkyl; or
    iv) R2 is hydroxyalkyl; or
    v) R2 is hydrogen; or
    vi) R2 is -CH2-C(OH)(CH3)2.
  3. A compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, of any one of claims 1 and 2, wherein R6 is hydrogen.
  4. A compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, of claim 1, wherein:
    i) R6 is hydrogen, -OR3 or -N(R3)(R4); or
    ii) only one of R1, R2 and R6 is H.
  5. A compound of formulae:
    Figure imgb0056
    Figure imgb0057
    Figure imgb0058
    Figure imgb0059
    Figure imgb0060
    Figure imgb0061
    Figure imgb0062
    or a pharmaceutically acceptable salt thereof; a tautomer thereof; or a pharmaceutically acceptable salt of the tautomer.
  6. A compound, , pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, according to any one of claims 1-5 for use in a method of:
    i) inducing interferon biosynthesis in a subject; or
    ii) modulating an immune response in a subject; or
    iii) inhibiting a kinase; or
    iv) inducing the production of TNF-α in a subject, wherein the compound optionally has an average steady-state drug concentration in the blood of less than 20 µM; or
    v) inducing an immune response in a subject, and optionally:
    i. wherein the immune response involves the production of cytokines; or
    ii. wherein the immune response involves modulation of TLR-7 and/or TLR-8; or
    iii. wherein the subject is suffering from a microbial infection; or
    iv. wherein the subject is suffering from a viral infection, such as a viral infection caused by the hepatitis C virus (HCV) or by the human immunodeficiency virus (HIV); or
    v. wherein the subject is suffering from abnormal cellular proliferation or cancer; or
    vi. wherein the subject is suffering from allergic diseases; or
    vii. wherein the subject is suffering from asthma; or
    viii. wherein the subject is suffering from precancerous lesions, such as actinic keratosis.
  7. A compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, according to claim 6 wherein the compound is administered topically.
  8. A compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, according to any one of the claims 1-5 for use in a method of:
    i) inducing an immune response in a subject, comprising: administering to the subject a compound according to any one of claims 1-5 and an antigen, wherein the compound induces an immune response to the antigen in the subj ect; or
    ii) enhancing the immune response to an antigen in a subject, comprising: administering to the subject a composition comprising a compound according to any one of claims 1-5 and an antigen, wherein the immune response to the antigen in the subject is enhanced.
  9. A pharmaceutical composition, comprising: the compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, of any one of claims 1-5 and a pharmaceutically acceptable excipient.
  10. A composition comprising the compound, pharmaceutically acceptable salt, tautomer, or a pharmaceutically acceptable salt of the tautomer, according to any one of claims 1-5 and an additional immunogenic composition or an antigen.
  11. The composition of claim 10, wherein the additional immunogenic composition comprises an antigen, for example wherein the antigen is:
    i) a bacterial antigen; or
    ii) a viral antigen, for example a virus selected from the group consisting of Hepatitis C virus, Human Immunodeficiency virus, Hepatitis B virus, Human Papiloma virus and Influenza virus.
  12. The composition of claim 10, wherein the antigen is:
    i) a bacterial antigen; or
    ii) a viral antigen, for example a virus selected from the group consisting of Hepatitis C virus, Human Immunodeficiency virus, Hepatitis B virus, Human Papiloma virus and Influenza virus.
  13. The composition according to any one of claims 9-12, further comprising an additional adjuvant, such as MF59.
  14. The composition according to any one of claims 12 or 13, further comprising poly(lactide-co-glycolide) (PLG).
EP11164995.0A 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators Not-in-force EP2357184B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US78554506P 2006-03-23 2006-03-23
EP07759315A EP2010537B1 (en) 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP07759315A Division EP2010537B1 (en) 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators
EP07759315.0 Division 2007-03-23

Publications (2)

Publication Number Publication Date
EP2357184A1 true EP2357184A1 (en) 2011-08-17
EP2357184B1 EP2357184B1 (en) 2015-02-25

Family

ID=38191164

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11164995.0A Not-in-force EP2357184B1 (en) 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators
EP07759315A Not-in-force EP2010537B1 (en) 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP07759315A Not-in-force EP2010537B1 (en) 2006-03-23 2007-03-23 Imidazoquinoxaline compounds as immunomodulators

Country Status (6)

Country Link
US (2) US8173657B2 (en)
EP (2) EP2357184B1 (en)
AT (1) ATE539079T1 (en)
CA (1) CA2646539A1 (en)
ES (2) ES2376492T3 (en)
WO (1) WO2007109813A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190104519A (en) * 2016-11-09 2019-09-10 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Methods and Compositions for Acquired Immune Regulation

Families Citing this family (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7612162B2 (en) 2004-09-21 2009-11-03 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Peptide analogs capable of enhancing stimulation of a glioma-specific CTL response
DK2121048T3 (en) 2007-02-19 2015-11-23 Marinepolymer Tech Inc Hemostatic compositions and therapeutic regimens
FR2921927B1 (en) 2007-10-03 2012-10-12 Univ De Montpellier 1 IMIDAZO® 1,2-A! QUINOXALINS AND DERIVATIVES FOR THE TREATMENT OF CANCER
WO2009089399A2 (en) * 2008-01-10 2009-07-16 Bausch & Lomb Incorporated Compositions comprising toll-like receptor or coreceptor antagonists and methods for ocular neuroprotection
CA2716706C (en) * 2008-03-03 2014-02-18 Irm Llc Compounds and compositions as tlr activity modulators
AU2010234849B2 (en) 2009-03-30 2017-06-22 Mount Sinai School Of Medicine Influenza virus vaccines and uses thereof
CN102762226A (en) * 2009-06-10 2012-10-31 诺华有限公司 Benzonaphthyridine-containing vaccines
EP2459585A1 (en) 2009-07-30 2012-06-06 Mount Sinai School of Medicine Influenza viruses and uses thereof
JP5988492B2 (en) * 2009-09-02 2016-09-07 ノバルティス アーゲー Immunogenic composition comprising a TLR activity modulator
TWI445708B (en) * 2009-09-02 2014-07-21 Irm Llc Compounds and compositions as tlr activity modulators
AU2011235220B2 (en) 2010-03-30 2016-03-10 Mount Sinai School Of Medicine Influenza virus vaccines and uses thereof
WO2011130646A1 (en) 2010-04-15 2011-10-20 Marine Polymer Technologies, Inc. Anti-bacterial applications of poly -n-acetylglucosamine nanofibers
ES2722799T3 (en) 2010-08-24 2019-08-16 Univ Pittsburgh Commonwealth Sys Higher Education Brain cancer vaccines based on the interleukin-13 receptor alpha 2 peptide
CA2810011A1 (en) 2010-09-01 2012-03-08 Novartis Ag Adsorption of immunopotentiators to insoluble metal salts
US20130287853A1 (en) 2010-11-06 2013-10-31 Marine Polymer Technologies, Inc. Compositions and methods for nanopolymer-based nucleic acid delivery
TR201811280T4 (en) * 2011-03-02 2018-08-27 Glaxosmithkline Biologicals Sa Mixed vaccines with low antigen and / or adjuvant doses.
CN103648279B (en) 2011-04-15 2017-05-24 海洋聚合物技术公司 Treatment of disease with poly-n-acety glucosamine nanofibers
WO2013043729A1 (en) 2011-09-20 2013-03-28 Mount Sinai School Of Medicine Influenza virus vaccines and uses thereof
US9375471B2 (en) 2012-03-08 2016-06-28 Glaxosmithkline Biologicals Sa Adjuvanted formulations of booster vaccines
ES2699817T3 (en) 2012-03-19 2019-02-12 Stemline Therapeutics Inc Methods to treat and control the state of a cancer
US10485858B2 (en) 2012-05-16 2019-11-26 Stemline Therapeutics, Inc. Cancer stem cell targeted cancer vaccines
JP6324961B2 (en) * 2012-09-06 2018-05-16 ノバルティス アーゲー Combination vaccine of serogroup B meningococcus and D / T / P
BR112015004817A2 (en) 2012-09-10 2017-07-04 Glycovaxyn Ag bioconjugates comprising modified antigens and their uses
BR112015008052A2 (en) 2012-10-12 2017-11-28 Glycovaxyn Ag host cell, methods for producing a glycoconjugate and for producing an unglycosylated protein, and, glycoconjugate
CN108641002A (en) 2012-12-18 2018-10-12 西奈山伊坎医学院 Influenza virus vaccine and application thereof
MX365331B (en) 2013-01-17 2019-05-29 Arsanis Biosciences Gmbh Mdr e. coli specific antibody.
WO2014159960A1 (en) 2013-03-14 2014-10-02 Icahn School Of Medicine At Mount Sinai Antibodies against influenza virus hemagglutinin and uses thereof
US9809801B2 (en) 2013-03-15 2017-11-07 Université De Genève Anti-mycobacterial vaccines
EP3077519B1 (en) 2013-12-03 2021-03-31 Hookipa Biotech GmbH Cmv vaccines
EA035991B9 (en) 2014-02-24 2020-10-21 Глаксосмитклайн Байолоджикалс С.А. Novel polysaccharide and uses thereof
EP3131577B1 (en) 2014-04-17 2020-04-22 GlaxoSmithKline Biologicals S.A. Modified host cells and uses thereof
AR104469A1 (en) 2014-08-08 2017-07-26 Glycovaxyn Ag GUEST CELLS MODIFIED FOR USE IN PRODUCTION OF BIOCONJUGATES
US9884866B2 (en) 2014-09-08 2018-02-06 Regents Of The University Of Minnesota Immunomodulators and immunomodulator conjugates
HUE051390T2 (en) 2014-11-13 2021-03-01 Univ Geneve Tri-segmented arenaviruses as vaccine vectors
FR3031105B1 (en) 2014-12-31 2018-04-06 Universite De Montpellier NOVEL IMIDAZO [1,2-A] QUINOXALINS AND DERIVATIVES FOR THE TREATMENT OF CANCER
AU2016209032A1 (en) 2015-01-23 2017-08-10 Icahn School Of Medicine At Mount Sinai Influenza virus vaccination regimens
CA2987155A1 (en) 2015-06-10 2016-12-15 Hookipa Biotech Ag Hpv vaccines
TWI715617B (en) 2015-08-24 2021-01-11 比利時商葛蘭素史密斯克藍生物品公司 Methods and compositions for immune protection against extra-intestinal pathogenic e. coli
WO2017037604A1 (en) * 2015-08-28 2017-03-09 Bheema Rao Paraselli Novel fyn kinase inhibitors
GB201518668D0 (en) 2015-10-21 2015-12-02 Glaxosmithkline Biolog Sa Immunogenic Comosition
CA3003557A1 (en) 2015-11-04 2017-05-11 Hookipa Biotech Ag Vaccines against hepatitis b virus
CN108697775B (en) 2015-11-12 2023-05-09 霍欧奇帕生物科技有限公司 Arenavirus particles as cancer vaccines
US10730871B2 (en) 2016-01-28 2020-08-04 Regents Of The University Of Minnesota Immunomodulators and immunomodulator conjugates
CN109804074B (en) 2016-05-18 2023-10-10 巴塞尔大学 Three-segment PICHINDE virus as vaccine vector
JP7237344B2 (en) 2016-06-15 2023-03-13 アイカーン スクール オブ メディシン アット マウント サイナイ Influenza virus hemagglutinin protein and uses thereof
GB201610599D0 (en) 2016-06-17 2016-08-03 Glaxosmithkline Biologicals Sa Immunogenic Composition
US11857352B2 (en) 2016-09-06 2024-01-02 The Research Foundation For The State University Of New York Positron imaging tomography imaging agent composition and method for bacterial infection
AR109621A1 (en) 2016-10-24 2018-12-26 Janssen Pharmaceuticals Inc FORMULATIONS OF VACCINES AGAINST GLUCOCONJUGADOS OF EXPEC
AU2017353443A1 (en) 2016-11-04 2019-05-02 Hookipa Biotech Gmbh Replication-deficient arenavirus particles and tri-segmented arenavirus particles as cancer vaccines
JP2020516601A (en) 2017-04-07 2020-06-11 ホオキパ バイオテック ジーエムビーエイチ Arenavirus particles for treating solid tumors
US11254733B2 (en) 2017-04-07 2022-02-22 Icahn School Of Medicine At Mount Sinai Anti-influenza B virus neuraminidase antibodies and uses thereof
GB201721576D0 (en) 2017-12-21 2018-02-07 Glaxosmithkline Biologicals Sa Hla antigens and glycoconjugates thereof
GB201721582D0 (en) 2017-12-21 2018-02-07 Glaxosmithkline Biologicals Sa S aureus antigens and immunogenic compositions
TW202003023A (en) 2018-03-12 2020-01-16 美商詹森藥物公司 Vaccines against urinary tract infections
WO2020120569A2 (en) 2018-12-12 2020-06-18 Glaxosmithkline Biologicals Sa Modified carrier proteins for o-linked glycosylation
JP7370391B2 (en) 2019-03-18 2023-10-27 ヤンセン ファーマシューティカルズ,インコーポレーテッド Bioconjugate of Escherichia coli O antigen polysaccharide, its production method and its use method
EA202192392A1 (en) 2019-03-18 2022-02-09 Янссен Фармасьютикалз, Инк. METHODS FOR PRODUCING E.COLI POLYSACCHARIDE O-ANTIGEN BIOCONJUGATES, THEIR COMPOSITIONS AND METHODS FOR THEIR APPLICATION
EP3770269A1 (en) 2019-07-23 2021-01-27 GlaxoSmithKline Biologicals S.A. Quantification of bioconjugate glycosylation
KR20220107166A (en) 2019-10-02 2022-08-02 얀센 백신스 앤드 프리벤션 비.브이. Staphylococcus Peptides and Methods of Use
WO2021069543A1 (en) 2019-10-08 2021-04-15 Luxembourg Institute Of Health (Lih) Inhibitor of dj-1 for use in treating immunoaging
AU2020377504A1 (en) 2019-11-07 2022-06-02 Universität Basel Arenaviruses as vectors
BR112022013720A2 (en) 2020-01-16 2022-10-11 Janssen Pharmaceuticals Inc MUTANT FIMH, COMPOSITIONS WITH IT AND ITS USE
CA3185719A1 (en) 2020-06-18 2021-12-23 Glaxosmithkline Biologicals Sa Shigella-tetravalent (shigella4v) bioconjugate
CA3190820A1 (en) 2020-09-17 2022-03-24 Janssen Pharmaceuticals, Inc. Multivalent vaccine compositions comprising specified ratios of e.coli o antigen polysaccharides and uses thereof
US20230197192A1 (en) 2020-11-06 2023-06-22 Amazon Technologies, Inc. Selecting neoantigens for personalized cancer vaccine
US20230416244A1 (en) * 2020-12-09 2023-12-28 Wellstat Therapeutics Corporation Imidazoquinoline compound having anti-inflammatory, antifugal, antiparasitic, and anticancer activity
AU2022207740A1 (en) 2021-01-12 2023-06-29 Janssen Pharmaceuticals, Inc. Fimh mutants, compositions therewith and use thereof
US20230074591A1 (en) 2021-01-19 2023-03-09 Amazon Technologies, Inc. A deep learning model for predicting tumor-specific neoantigen mhc class i or class ii immunogenicity
CN117157713A (en) 2021-02-05 2023-12-01 亚马逊科技公司 Ranking neoantigens for personalized cancer vaccine
JP2024512462A (en) 2021-03-15 2024-03-19 アマゾン テクノロジーズ インコーポレイテッド Methods to optimize tumor vaccine antigen coverage for heterogeneous malignancies
BR112023019365A2 (en) 2021-03-23 2023-12-26 Hookipa Biotech Gmbh ARENAVIRUS S-SEGMENT, CDNA, DNA EXPRESSION VECTOR, HOST CELL, THREE-SEGMENTED ARENAVIRUS PARTICLE, METHOD FOR GENERATING A THREE-SEGMENTED ARENAVIRUS PARTICLE, PHARMACEUTICAL COMPOSITION, METHOD FOR TREATING PROSTATE CANCER AND KIT
KR20230164108A (en) 2021-04-01 2023-12-01 얀센 파마슈티칼즈, 인코포레이티드 Production of E. coli O18 bioconjugate
WO2022238546A1 (en) 2021-05-13 2022-11-17 Hookipa Biotech Gmbh Arenaviruses as vectors
US20230173046A1 (en) 2021-05-27 2023-06-08 Amazon Technologies, Inc. Multicomponent chemical composition of a peptide-based neoantigen vaccine
TW202334403A (en) 2021-11-08 2023-09-01 奧地利商霍歐奇帕生物科技有限公司 Modified arenavirus particles expressing mutant kras, mutated cancer driver gene, or tumor-associated antigen as cancer immunotherapies
WO2023152116A1 (en) 2022-02-08 2023-08-17 Hookipa Biotech Gmbh Combination therapy with arenavirus particles and immune checkpoint modulators or cytokines
WO2023175044A1 (en) 2022-03-15 2023-09-21 Institut Pasteur Multivalent mopevac-based immunogenic composition for vaccination against new world arenaviruses and therapeutic use(s) thereof
WO2023178229A1 (en) 2022-03-16 2023-09-21 Amazon Technologies, Inc. Monitoring circulating tumor dna to improve subclone penetration of follow-up neoantigen cancer vaccines
WO2024015702A1 (en) 2022-07-15 2024-01-18 Amazon Technologies, Inc. Personalized longitudinal analysis of circulating material to monitor and adapt neoantigen cancer vaccines

Citations (141)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0109942A2 (en) 1982-10-18 1984-05-30 Bror Morein Immunogenic protein or peptide complex, method of producing said complex and the use thereof as an immune stimulant and as a vaccine
US4689338A (en) 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
US4806352A (en) 1986-04-15 1989-02-21 Ribi Immunochem Research Inc. Immunological lipid emulsion adjuvant
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
EP0372501A2 (en) 1988-12-07 1990-06-13 BEHRINGWERKE Aktiengesellschaft Synthetic antigens, method for their preparation and their use
EP0378881A1 (en) 1989-01-17 1990-07-25 ENIRICERCHE S.p.A. Synthetic peptides and their use as universal carriers for the preparation of immunogenic conjugates suitable for the development of synthetic vaccines
WO1990014837A1 (en) 1989-05-25 1990-12-13 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
WO1991002062A2 (en) 1989-08-04 1991-02-21 Triton Biosciences, Inc. C-erbb-2 external domain: gp75
US5011828A (en) 1985-11-15 1991-04-30 Michael Goodman Immunostimulating guanine derivatives, compositions and methods
EP0427347A1 (en) 1989-11-10 1991-05-15 ENIRICERCHE S.p.A. Synthetic peptides useful as universal carriers for the preparation of immunogenic conjugates and their use in the development of synthetic vaccines
US5026543A (en) 1987-03-17 1991-06-25 Akzo N.V. Adjuvant mixture
US5026546A (en) 1986-12-19 1991-06-25 Duphar International Research B.V. Stabilized adjuvant suspension comprising dimethyl dioctadecyl ammonium bromide
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
EP0471177A2 (en) 1990-08-13 1992-02-19 American Cyanamid Company Filamentous hemagglutinin of bordetella pertussis as a carrier molecule for conjugate vaccines
EP0477508A1 (en) 1990-09-28 1992-04-01 American Cyanamid Company Improved oligosaccharide conjugate vaccines
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
WO1993017712A2 (en) 1992-03-06 1993-09-16 Biocine Spa Conjugates formed from heat shock proteins and oligo- or polysaccharides
WO1993018150A1 (en) 1992-03-02 1993-09-16 Biocine S.P.A. Helicobacter pylori proteins useful for vaccines and diagnostics
WO1993019780A1 (en) 1992-03-27 1993-10-14 Smithkline Beecham Biologicals (S.A.) Hepatitis vaccines containing 3-o-deacylated monophoshoryl lipid a
US5266575A (en) 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
US5268376A (en) 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
WO1994000153A1 (en) 1992-06-25 1994-01-06 Smithkline Beecham Biologicals (S.A.) Vaccine composition containing adjuvants
WO1994021292A1 (en) 1993-03-23 1994-09-29 Smithkline Beecham Biologicals (S.A.) Vaccine compositions containing 3-o deacylated monophosphoryl lipid a
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5360897A (en) 1981-08-31 1994-11-01 The University Of Rochester Immunogenic conjugates of streptococcus pneumonial capsular polymer and toxin or in toxiad
EP0626169A2 (en) 1988-08-25 1994-11-30 The Liposome Company, Inc. A dosage form comprising an antigen and a salt form of an organic acid derivative of a sterol
US5378814A (en) 1986-06-17 1995-01-03 Chiron Corporation Hepatitis delta viral polypeptides
US5389640A (en) 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
WO1995017211A1 (en) 1993-12-22 1995-06-29 Biocine S.P.A. Non-toxic mucosal adjuvant
WO1995017210A1 (en) 1993-12-23 1995-06-29 Smithkline Beecham Biologicals (S.A.) Vaccines
US5482936A (en) 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
US5494916A (en) 1993-07-15 1996-02-27 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]pyridin-4-amines
WO1996011711A1 (en) 1994-10-12 1996-04-25 Iscotec Ab Saponin preparations and use thereof in iscoms
WO1996026741A1 (en) 1995-02-25 1996-09-06 Smithkline Beeecham Biologicals S.A. Hepatitis b vaccine
WO1996029412A1 (en) 1995-03-17 1996-09-26 Biochem Vaccines Inc. Proteinase k resistant surface protein of neisseria meningitidis
WO1996030514A1 (en) 1995-03-31 1996-10-03 University Of Washington Intracellular domain of the her-2/neu protein for prevention or treatment of malignancies
WO1996033739A1 (en) 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
WO1997019079A1 (en) * 1995-11-24 1997-05-29 Biomedica Foscama Industria Chimico-Farmaceutica S.P.A. IMIDAZO[1,2-a]QUINOXALIN-4-AMINES ACTIVE AS ADENOSINE ANTAGONISTS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS THEREOF
US5658731A (en) 1990-04-09 1997-08-19 Europaisches Laboratorium Fur Molekularbiologie 2'-O-alkylnucleotides as well as polymers which contain such nucleotides
WO1997037026A1 (en) 1996-04-02 1997-10-09 Smithkline Beecham Corporation Novel compounds
WO1997043303A1 (en) 1996-05-14 1997-11-20 Smithkline Beecham Corporation Novel compounds
US5693522A (en) 1991-11-29 1997-12-02 Chiron Viagene, Inc. Anti-cancer immunotherapeutics
WO1998004702A2 (en) 1996-07-26 1998-02-05 Chiron Behring Gmbh & Co. Proteins, in particular membrane proteins, of helicobacter pylori, their preparation and use
EP0835318A2 (en) 1995-06-29 1998-04-15 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccines against hepatitis c
WO1998018931A2 (en) 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae polynucleotides and sequences
WO1998040100A1 (en) 1997-03-10 1998-09-17 Ottawa Civic Loeb Research Institute USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE AS AN ADJUVANT
WO1998042375A1 (en) 1997-03-21 1998-10-01 Chiron Corporation Detoxified mutants of bacterial adp-ribosylating toxins as parenteral adjuvants
WO1998042721A1 (en) 1997-03-24 1998-10-01 Andrew Lees Uronium salt conjugate vaccines
WO1998057659A1 (en) 1997-06-14 1998-12-23 Smithkline Beecham Biologicals S.A. Adjuvant compositions for vaccines
WO1998058668A2 (en) 1997-06-20 1998-12-30 Microbiological Research Authority Bordetella pertussis antigens as carriers in vaccinating conjugates and oral vaccines comprising bordetella pertussis fimbriae
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
WO1999009845A1 (en) * 1997-08-25 1999-03-04 Bristol-Myers Squibb Company Imidazoquinoxaline protein tyrosine kinase inhibitors
WO1999011241A1 (en) 1997-09-05 1999-03-11 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
WO1999024578A2 (en) 1997-11-06 1999-05-20 Chiron S.P.A. Neisserial antigens
WO1999027105A2 (en) 1997-11-21 1999-06-03 Genset Chlamydia pneumoniae genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
WO1999027960A1 (en) 1997-11-28 1999-06-10 West Pharmaceutical Services Vaccine compositions for mucosal administration comprising chitosan
WO1999028475A2 (en) 1997-11-28 1999-06-10 Genset Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
WO1999029693A1 (en) 1997-12-11 1999-06-17 Minnesota Mining And Manufacturing Company Imidazonaphthyridines and their use in inducing cytokine biosynthesis
US5916588A (en) 1984-04-12 1999-06-29 The Liposome Company, Inc. Peptide-containing liposomes, immunogenic liposomes and methods of preparation and use
WO1999036544A2 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
WO1999053310A1 (en) 1998-04-08 1999-10-21 Chiron S.P.A. Antigen
WO1999052549A1 (en) 1998-04-09 1999-10-21 Smithkline Beecham Biologicals S.A. Adjuvant compositions
WO1999057280A2 (en) 1998-05-01 1999-11-11 Chiron Corporation Neisseria meningitidis antigens and compositions
WO1999058562A2 (en) 1998-05-12 1999-11-18 Smithkline Beecham Biologicals S.A. Novel compounds
WO1999062923A2 (en) 1998-06-05 1999-12-09 Dynavax Technologies Corporation Immunostimulatory oligonucleotides with modified bases and methods of use thereof
US6015567A (en) 1989-05-19 2000-01-18 Genentech, Inc. HER2 extracellular domain
WO2000007621A2 (en) 1998-08-05 2000-02-17 Smithkline Beecham Biologicals S.A. Vaccine comprising an iscom consisting of sterol and saponin which is free of additional detergent
WO2000022430A2 (en) 1998-10-09 2000-04-20 Chiron Corporation Neisseria genomic sequences and methods of their use
WO2000023105A2 (en) 1998-10-16 2000-04-27 Smithkline Beecham Biologicals S.A. Adjuvant systems and vaccines
WO2000027994A2 (en) 1998-11-12 2000-05-18 The Regents Of The University Of California Chlamydia pneumoniae genome sequence
WO2000037494A2 (en) 1998-12-18 2000-06-29 Chiron S.P.A. Chlamydia trachomatis antigens
US6083505A (en) 1992-04-16 2000-07-04 3M Innovative Properties Company 1H-imidazo[4,5-C]quinolin-4-amines as vaccine adjuvants
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
US6110929A (en) 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
WO2000056360A2 (en) 1999-03-19 2000-09-28 Smithkline Beecham Biologicals S.A. Vaccine against antigens from bacteriae
WO2000067161A2 (en) 1999-05-04 2000-11-09 Grant Lee H Method and apparatus for categorizing and retrieving network pages and sites
WO2001008636A2 (en) 1999-08-03 2001-02-08 The Ohio State University Polypeptides and polynucleotides for enhancing immune reactivity to her-2 protein
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO2001021152A1 (en) 1999-09-24 2001-03-29 Smithkline Beecham Biologicals S.A. Adjuvant comprising a polyxyethylene alkyl ether or ester and at least one nonionic surfactant
WO2001021207A2 (en) 1999-09-24 2001-03-29 Smithkline Beecham Biologicals S.A. Use of combination of polyoxyethylene sorbitan ester and octoxynol as adjuvant and its use in vaccines
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO2001037869A1 (en) 1999-11-19 2001-05-31 Csl Limited Vaccine compositions
WO2001052885A1 (en) 2000-01-17 2001-07-26 Chiron Spa Outer membrane vesicle (omv) vaccine comprising n. meningitidis serogroup b outer membrane proteins
US6284772B1 (en) 1998-09-30 2001-09-04 The United States Of America As Represented By The Secretary Of The Army Indolo[2,1-B] quinazole-6,12-dione antimalarial compounds and methods of treating malaria therewith
WO2001095935A1 (en) 2000-01-20 2001-12-20 Ottawa Health Research Institute Immunostimulatory nucleic acids for inducing a th2 immune response
US6333164B1 (en) 1996-09-04 2001-12-25 Takara Shuzo Co., Ltd. Fungal antigens and process for producing the same
WO2002002606A2 (en) 2000-07-03 2002-01-10 Chiron S.P.A. Immunisation against chlamydia pneumoniae
US20020007173A1 (en) 1997-07-10 2002-01-17 Kundig Thomas M. Method of inducing a CTL response
WO2002018595A2 (en) 2000-08-28 2002-03-07 Aventis Pasteur Limited Moraxella polypeptides and corresponding dna fragments and uses thereof
WO2002018383A2 (en) 2000-09-01 2002-03-07 Chiron Corporation Aza heterocyclic derivatives and their therapeutic use
WO2002026757A2 (en) 2000-09-26 2002-04-04 Hybridon, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
WO2002034771A2 (en) 2000-10-27 2002-05-02 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
WO2002079243A2 (en) 2001-02-12 2002-10-10 Chiron Srl. Gonococcal proteins and nucleic acids
WO2002085905A1 (en) 2001-04-17 2002-10-31 Sumitomo Pharmaceuticals Company, Limited Novel adenine derivatives
WO2002094851A2 (en) 2001-05-18 2002-11-28 The Government Of The United States Of America, Asrepresented By The Secretary, Department Of Healthand Human Services, Centers For Disease Control And Prevention, Technology Transfer Office Peptide vaccines against group a streptococci
WO2003002065A2 (en) 2001-06-29 2003-01-09 Chiron Corporation Hcv e1e2 vaccine compositions
US20030022898A1 (en) * 2000-10-03 2003-01-30 Burke James R. Methods of treating inflammatory and immune diseases using inhibitors of IkappaB kinase (IKK)
WO2003011223A2 (en) 2001-07-31 2003-02-13 Eisai Co., Ltd. Immunomodulatory compounds and methods of use thereof
WO2003024481A2 (en) 2001-09-14 2003-03-27 Cytos Biotechnology Ag Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
WO2003024480A2 (en) 2001-09-14 2003-03-27 Cytos Biotechnology Ag In vivo activation of antigen presenting cells for enhancement of immune responses induced by virus like particles
WO2003035836A2 (en) 2001-10-24 2003-05-01 Hybridon Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5' ends
WO2003043572A2 (en) 2001-11-16 2003-05-30 3M Innovative Properties Company Methods and compositions related to irm compounds and toll-like receptor pathways
WO2003049762A2 (en) 2001-12-12 2003-06-19 Chiron Srl. Immunisation against chlamydia trachomatis
US6605617B2 (en) 2000-09-11 2003-08-12 Chiron Corporation Quinolinone derivatives
WO2003068811A2 (en) 2002-02-13 2003-08-21 Chiron Srl Cytotoxic t-cell epitopes from chlamydia
US6630161B1 (en) 1998-05-07 2003-10-07 Ribi Immunochem Research, Inc. Adjuvant composition and methods for its use
WO2003082272A1 (en) 2002-03-29 2003-10-09 Chiron Corporation Substituted benzazoles and use thereof as raf kinase inhibitors
WO2003093306A2 (en) 2002-05-02 2003-11-13 Chir0N Srl Nucleic acids and proteins from streptococcus groups a & b
WO2003097641A2 (en) 2002-05-21 2003-11-27 Novartis Ag 1h-imidazo[4,5-c] quinoline derivatives in the treatment of protein kinase dependent diseases
US6656938B2 (en) 2000-12-08 2003-12-02 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660735B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660747B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6664260B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Heterocyclic ether substituted imidazoquinolines
US6664265B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6664264B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6667312B2 (en) 2000-12-08 2003-12-23 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6677349B1 (en) 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6677348B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6677347B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
WO2004005473A2 (en) 2002-07-02 2004-01-15 Chiron Corporation Hcv fusion proteins with modified ns3 domains
US6699703B1 (en) 1997-07-02 2004-03-02 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
WO2004018455A1 (en) 2002-08-23 2004-03-04 Chiron Corporation Pyrrole based inhibitors of glycogen synthase kinase 3
WO2004041157A2 (en) 2002-09-13 2004-05-21 Chiron Corporation Group b streptococcus vaccine
US6743920B2 (en) 2002-05-29 2004-06-01 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
US6756361B1 (en) 1997-10-14 2004-06-29 Nabi Enterococcus antigens and vaccines
WO2004060308A2 (en) 2002-12-27 2004-07-22 Chiron Corporation Thiosemicarbazones as anti-virals and immunopotentiators
WO2004064715A2 (en) 2003-01-23 2004-08-05 M N L Pharma Limited Polyhydroxylated pyrrolizidine
WO2004064759A2 (en) 2003-01-21 2004-08-05 Chiron Corporation Use of tryptanthrin compounds for immune potentiation
WO2004071459A2 (en) 2003-02-13 2004-08-26 3M Innovative Properties Company Methods and compositions related to irm compounds and toll-like receptor 8
WO2004076677A2 (en) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Monoclonal antibody production by ebv transformation of b cells
US6800624B2 (en) 1999-06-10 2004-10-05 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
WO2004087153A2 (en) 2003-03-28 2004-10-14 Chiron Corporation Use of organic compounds for immunopotentiation
WO2004092360A2 (en) 2003-04-10 2004-10-28 Chiron Corporation The severe acute respiratory syndrome coronavirus
WO2005002619A2 (en) 2003-06-26 2005-01-13 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
WO2005025614A2 (en) 2003-09-15 2005-03-24 Glaxo Group Limited Improvements in vaccination
US20050070556A1 (en) 2001-11-27 2005-03-31 Anadys Pharmaceuticals, Inc. 3-B-D-ribofuranosylthiazolo [4,5-d] pyridimine nucleosides and uses thereof
WO2005032582A2 (en) 2003-07-31 2005-04-14 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
US6924271B2 (en) 2001-11-27 2005-08-02 Anadys Pharmaceuticals, Inc. 3-β-D-ribofuranosylthiazolo[4-5-d]pyridimine nucleosides and uses thereof
US20050215517A1 (en) 1999-01-14 2005-09-29 Rossignol Daniel P Use of an anti-endotoxin drug in the prevention and treatment of disease

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US666747A (en) * 1900-11-05 1901-01-29 American Pneumatic Service Co Carrier for pneumatic-despatch tubes.
GB263335A (en) 1926-02-11 1926-12-30 Cannon Iron Founders Ltd Improvements relating to atmospheric gas burners
US4578458A (en) * 1983-03-23 1986-03-25 Brigham And Women's Hospital Mucoid exopolysaccharide vaccine against Pseudomonas aeruginosa
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
US6884435B1 (en) * 1997-01-30 2005-04-26 Chiron Corporation Microparticles with adsorbent surfaces, methods of making same, and uses thereof
US6180650B1 (en) * 1999-04-23 2001-01-30 Merck Frosst Canada & Co. Heterosubstituted pyridine derivatives as PDE 4 inhibitors
US6861410B1 (en) * 2002-03-21 2005-03-01 Chiron Corporation Immunological adjuvant compositions
GB2388929B (en) * 2002-05-23 2005-05-18 Advanced Risc Mach Ltd Handling of a multi-access instruction in a data processing apparatus
ES2596553T3 (en) * 2003-06-02 2017-01-10 Glaxosmithkline Biologicals Sa Immunogenic compositions based on microparticles comprising adsorbed toxoid and an antigen containing a polysaccharide

Patent Citations (160)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5360897A (en) 1981-08-31 1994-11-01 The University Of Rochester Immunogenic conjugates of streptococcus pneumonial capsular polymer and toxin or in toxiad
EP0109942A2 (en) 1982-10-18 1984-05-30 Bror Morein Immunogenic protein or peptide complex, method of producing said complex and the use thereof as an immune stimulant and as a vaccine
US4689338A (en) 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
US5916588A (en) 1984-04-12 1999-06-29 The Liposome Company, Inc. Peptide-containing liposomes, immunogenic liposomes and methods of preparation and use
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
US5011828A (en) 1985-11-15 1991-04-30 Michael Goodman Immunostimulating guanine derivatives, compositions and methods
US4806352A (en) 1986-04-15 1989-02-21 Ribi Immunochem Research Inc. Immunological lipid emulsion adjuvant
US5378814A (en) 1986-06-17 1995-01-03 Chiron Corporation Hepatitis delta viral polypeptides
US5026546A (en) 1986-12-19 1991-06-25 Duphar International Research B.V. Stabilized adjuvant suspension comprising dimethyl dioctadecyl ammonium bromide
US5026543A (en) 1987-03-17 1991-06-25 Akzo N.V. Adjuvant mixture
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
EP0626169A2 (en) 1988-08-25 1994-11-30 The Liposome Company, Inc. A dosage form comprising an antigen and a salt form of an organic acid derivative of a sterol
EP0372501A2 (en) 1988-12-07 1990-06-13 BEHRINGWERKE Aktiengesellschaft Synthetic antigens, method for their preparation and their use
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
EP0378881A1 (en) 1989-01-17 1990-07-25 ENIRICERCHE S.p.A. Synthetic peptides and their use as universal carriers for the preparation of immunogenic conjugates suitable for the development of synthetic vaccines
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
US6015567A (en) 1989-05-19 2000-01-18 Genentech, Inc. HER2 extracellular domain
US6451325B1 (en) 1989-05-25 2002-09-17 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
WO1990014837A1 (en) 1989-05-25 1990-12-13 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
US6299884B1 (en) 1989-05-25 2001-10-09 Chiron Corporation Adjuvant formulation comprising a submicron oil droplet emulsion
WO1991002062A2 (en) 1989-08-04 1991-02-21 Triton Biosciences, Inc. C-erbb-2 external domain: gp75
EP0427347A1 (en) 1989-11-10 1991-05-15 ENIRICERCHE S.p.A. Synthetic peptides useful as universal carriers for the preparation of immunogenic conjugates and their use in the development of synthetic vaccines
US5658731A (en) 1990-04-09 1997-08-19 Europaisches Laboratorium Fur Molekularbiologie 2'-O-alkylnucleotides as well as polymers which contain such nucleotides
EP0471177A2 (en) 1990-08-13 1992-02-19 American Cyanamid Company Filamentous hemagglutinin of bordetella pertussis as a carrier molecule for conjugate vaccines
US5306492A (en) 1990-09-28 1994-04-26 American Cyanamid Company Oligosaccharide conjugate vaccines
EP0477508A1 (en) 1990-09-28 1992-04-01 American Cyanamid Company Improved oligosaccharide conjugate vaccines
US5389640A (en) 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5268376A (en) 1991-09-04 1993-12-07 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5346905A (en) 1991-09-04 1994-09-13 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo-[4,5-C]quinolin-4-amines
US5525612A (en) 1991-09-04 1996-06-11 Minnesota Mining And Manufacturing Company 1-substituted 1H-imidazo-[4,5-c]quinolin-4-amines
US5266575A (en) 1991-11-06 1993-11-30 Minnesota Mining And Manufacturing Company 2-ethyl 1H-imidazo[4,5-ciquinolin-4-amines
US5693522A (en) 1991-11-29 1997-12-02 Chiron Viagene, Inc. Anti-cancer immunotherapeutics
WO1993018150A1 (en) 1992-03-02 1993-09-16 Biocine S.P.A. Helicobacter pylori proteins useful for vaccines and diagnostics
WO1993017712A2 (en) 1992-03-06 1993-09-16 Biocine Spa Conjugates formed from heat shock proteins and oligo- or polysaccharides
WO1993019780A1 (en) 1992-03-27 1993-10-14 Smithkline Beecham Biologicals (S.A.) Hepatitis vaccines containing 3-o-deacylated monophoshoryl lipid a
US6083505A (en) 1992-04-16 2000-07-04 3M Innovative Properties Company 1H-imidazo[4,5-C]quinolin-4-amines as vaccine adjuvants
WO1994000153A1 (en) 1992-06-25 1994-01-06 Smithkline Beecham Biologicals (S.A.) Vaccine composition containing adjuvants
EP0761231A1 (en) 1992-06-25 1997-03-12 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccine composition containing adjuvants
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5846538A (en) 1993-03-17 1998-12-08 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis and treatment of malignancies in which the her-2/neu oncogene is associated
EP0689454A1 (en) 1993-03-23 1996-01-03 Smithkline Beecham Biolog Vaccine compositions containing 3-o deacylated monophosphoryl lipid a
WO1994021292A1 (en) 1993-03-23 1994-09-29 Smithkline Beecham Biologicals (S.A.) Vaccine compositions containing 3-o deacylated monophosphoryl lipid a
US5494916A (en) 1993-07-15 1996-02-27 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]pyridin-4-amines
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
WO1995017211A1 (en) 1993-12-22 1995-06-29 Biocine S.P.A. Non-toxic mucosal adjuvant
EP0735898A1 (en) 1993-12-23 1996-10-09 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccines
WO1995017210A1 (en) 1993-12-23 1995-06-29 Smithkline Beecham Biologicals (S.A.) Vaccines
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
WO1996011711A1 (en) 1994-10-12 1996-04-25 Iscotec Ab Saponin preparations and use thereof in iscoms
US5482936A (en) 1995-01-12 1996-01-09 Minnesota Mining And Manufacturing Company Imidazo[4,5-C]quinoline amines
WO1996026741A1 (en) 1995-02-25 1996-09-06 Smithkline Beeecham Biologicals S.A. Hepatitis b vaccine
WO1996029412A1 (en) 1995-03-17 1996-09-26 Biochem Vaccines Inc. Proteinase k resistant surface protein of neisseria meningitidis
WO1996030514A1 (en) 1995-03-31 1996-10-03 University Of Washington Intracellular domain of the her-2/neu protein for prevention or treatment of malignancies
WO1996033739A1 (en) 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
EP0835318A2 (en) 1995-06-29 1998-04-15 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccines against hepatitis c
WO1997019079A1 (en) * 1995-11-24 1997-05-29 Biomedica Foscama Industria Chimico-Farmaceutica S.P.A. IMIDAZO[1,2-a]QUINOXALIN-4-AMINES ACTIVE AS ADENOSINE ANTAGONISTS, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS THEREOF
WO1997037026A1 (en) 1996-04-02 1997-10-09 Smithkline Beecham Corporation Novel compounds
WO1997043303A1 (en) 1996-05-14 1997-11-20 Smithkline Beecham Corporation Novel compounds
WO1998004702A2 (en) 1996-07-26 1998-02-05 Chiron Behring Gmbh & Co. Proteins, in particular membrane proteins, of helicobacter pylori, their preparation and use
US6333164B1 (en) 1996-09-04 2001-12-25 Takara Shuzo Co., Ltd. Fungal antigens and process for producing the same
WO1998018930A2 (en) 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
WO1998018931A2 (en) 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae polynucleotides and sequences
WO1998040100A1 (en) 1997-03-10 1998-09-17 Ottawa Civic Loeb Research Institute USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE AS AN ADJUVANT
WO1998042375A1 (en) 1997-03-21 1998-10-01 Chiron Corporation Detoxified mutants of bacterial adp-ribosylating toxins as parenteral adjuvants
WO1998042721A1 (en) 1997-03-24 1998-10-01 Andrew Lees Uronium salt conjugate vaccines
WO1998057659A1 (en) 1997-06-14 1998-12-23 Smithkline Beecham Biologicals S.A. Adjuvant compositions for vaccines
WO1998058668A2 (en) 1997-06-20 1998-12-30 Microbiological Research Authority Bordetella pertussis antigens as carriers in vaccinating conjugates and oral vaccines comprising bordetella pertussis fimbriae
US6699703B1 (en) 1997-07-02 2004-03-02 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US6800744B1 (en) 1997-07-02 2004-10-05 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US20020007173A1 (en) 1997-07-10 2002-01-17 Kundig Thomas M. Method of inducing a CTL response
WO1999009845A1 (en) * 1997-08-25 1999-03-04 Bristol-Myers Squibb Company Imidazoquinoxaline protein tyrosine kinase inhibitors
WO1999011241A1 (en) 1997-09-05 1999-03-11 Smithkline Beecham Biologicals S.A. Oil in water emulsions containing saponins
US6756361B1 (en) 1997-10-14 2004-06-29 Nabi Enterococcus antigens and vaccines
WO1999024578A2 (en) 1997-11-06 1999-05-20 Chiron S.P.A. Neisserial antigens
WO1999027105A2 (en) 1997-11-21 1999-06-03 Genset Chlamydia pneumoniae genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
WO1999028475A2 (en) 1997-11-28 1999-06-10 Genset Chlamydia trachomatis genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
WO1999027960A1 (en) 1997-11-28 1999-06-10 West Pharmaceutical Services Vaccine compositions for mucosal administration comprising chitosan
WO1999029693A1 (en) 1997-12-11 1999-06-17 Minnesota Mining And Manufacturing Company Imidazonaphthyridines and their use in inducing cytokine biosynthesis
WO1999036544A2 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
WO1999053310A1 (en) 1998-04-08 1999-10-21 Chiron S.P.A. Antigen
WO1999052549A1 (en) 1998-04-09 1999-10-21 Smithkline Beecham Biologicals S.A. Adjuvant compositions
WO1999057280A2 (en) 1998-05-01 1999-11-11 Chiron Corporation Neisseria meningitidis antigens and compositions
US6630161B1 (en) 1998-05-07 2003-10-07 Ribi Immunochem Research, Inc. Adjuvant composition and methods for its use
WO1999058562A2 (en) 1998-05-12 1999-11-18 Smithkline Beecham Biologicals S.A. Novel compounds
WO1999062923A2 (en) 1998-06-05 1999-12-09 Dynavax Technologies Corporation Immunostimulatory oligonucleotides with modified bases and methods of use thereof
US6809203B2 (en) 1998-07-28 2004-10-26 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-C]-quinolin-4-amines and analogs thereof
US6703402B2 (en) 1998-07-28 2004-03-09 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US6627640B2 (en) 1998-07-28 2003-09-30 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US6110929A (en) 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
US6440992B1 (en) 1998-07-28 2002-08-27 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
WO2000007621A2 (en) 1998-08-05 2000-02-17 Smithkline Beecham Biologicals S.A. Vaccine comprising an iscom consisting of sterol and saponin which is free of additional detergent
US6284772B1 (en) 1998-09-30 2001-09-04 The United States Of America As Represented By The Secretary Of The Army Indolo[2,1-B] quinazole-6,12-dione antimalarial compounds and methods of treating malaria therewith
WO2000022430A2 (en) 1998-10-09 2000-04-20 Chiron Corporation Neisseria genomic sequences and methods of their use
WO2000023105A2 (en) 1998-10-16 2000-04-27 Smithkline Beecham Biologicals S.A. Adjuvant systems and vaccines
WO2000027994A2 (en) 1998-11-12 2000-05-18 The Regents Of The University Of California Chlamydia pneumoniae genome sequence
WO2000037494A2 (en) 1998-12-18 2000-06-29 Chiron S.P.A. Chlamydia trachomatis antigens
US20050215517A1 (en) 1999-01-14 2005-09-29 Rossignol Daniel P Use of an anti-endotoxin drug in the prevention and treatment of disease
WO2000056360A2 (en) 1999-03-19 2000-09-28 Smithkline Beecham Biologicals S.A. Vaccine against antigens from bacteriae
WO2000067161A2 (en) 1999-05-04 2000-11-09 Grant Lee H Method and apparatus for categorizing and retrieving network pages and sites
US6800624B2 (en) 1999-06-10 2004-10-05 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
WO2001008636A2 (en) 1999-08-03 2001-02-08 The Ohio State University Polypeptides and polynucleotides for enhancing immune reactivity to her-2 protein
WO2001021152A1 (en) 1999-09-24 2001-03-29 Smithkline Beecham Biologicals S.A. Adjuvant comprising a polyxyethylene alkyl ether or ester and at least one nonionic surfactant
WO2001021207A2 (en) 1999-09-24 2001-03-29 Smithkline Beecham Biologicals S.A. Use of combination of polyoxyethylene sorbitan ester and octoxynol as adjuvant and its use in vaccines
WO2001037869A1 (en) 1999-11-19 2001-05-31 Csl Limited Vaccine compositions
WO2001052885A1 (en) 2000-01-17 2001-07-26 Chiron Spa Outer membrane vesicle (omv) vaccine comprising n. meningitidis serogroup b outer membrane proteins
WO2001095935A1 (en) 2000-01-20 2001-12-20 Ottawa Health Research Institute Immunostimulatory nucleic acids for inducing a th2 immune response
WO2002002606A2 (en) 2000-07-03 2002-01-10 Chiron S.P.A. Immunisation against chlamydia pneumoniae
WO2002018595A2 (en) 2000-08-28 2002-03-07 Aventis Pasteur Limited Moraxella polypeptides and corresponding dna fragments and uses thereof
WO2002018383A2 (en) 2000-09-01 2002-03-07 Chiron Corporation Aza heterocyclic derivatives and their therapeutic use
US6605617B2 (en) 2000-09-11 2003-08-12 Chiron Corporation Quinolinone derivatives
WO2002026757A2 (en) 2000-09-26 2002-04-04 Hybridon, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
US20030022898A1 (en) * 2000-10-03 2003-01-30 Burke James R. Methods of treating inflammatory and immune diseases using inhibitors of IkappaB kinase (IKK)
WO2002034771A2 (en) 2000-10-27 2002-05-02 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
US6677347B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US6664260B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Heterocyclic ether substituted imidazoquinolines
US6667312B2 (en) 2000-12-08 2003-12-23 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664264B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Thioether substituted imidazoquinolines
US6664265B2 (en) 2000-12-08 2003-12-16 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6683088B2 (en) 2000-12-08 2004-01-27 3M Innovative Properties Company Sulfonamido ether substituted imidazoquinolines
US6677348B2 (en) 2000-12-08 2004-01-13 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
US6656938B2 (en) 2000-12-08 2003-12-02 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660735B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Urea substituted imidazoquinoline ethers
US6660747B2 (en) 2000-12-08 2003-12-09 3M Innovative Properties Company Amido ether substituted imidazoquinolines
US6670372B2 (en) 2000-12-08 2003-12-30 3M Innovative Properties Company Aryl ether substituted imidazoquinolines
WO2002079243A2 (en) 2001-02-12 2002-10-10 Chiron Srl. Gonococcal proteins and nucleic acids
WO2002085905A1 (en) 2001-04-17 2002-10-31 Sumitomo Pharmaceuticals Company, Limited Novel adenine derivatives
WO2002094851A2 (en) 2001-05-18 2002-11-28 The Government Of The United States Of America, Asrepresented By The Secretary, Department Of Healthand Human Services, Centers For Disease Control And Prevention, Technology Transfer Office Peptide vaccines against group a streptococci
WO2003002065A2 (en) 2001-06-29 2003-01-09 Chiron Corporation Hcv e1e2 vaccine compositions
WO2003011223A2 (en) 2001-07-31 2003-02-13 Eisai Co., Ltd. Immunomodulatory compounds and methods of use thereof
WO2003024480A2 (en) 2001-09-14 2003-03-27 Cytos Biotechnology Ag In vivo activation of antigen presenting cells for enhancement of immune responses induced by virus like particles
WO2003024481A2 (en) 2001-09-14 2003-03-27 Cytos Biotechnology Ag Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
WO2003035836A2 (en) 2001-10-24 2003-05-01 Hybridon Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5' ends
WO2003043572A2 (en) 2001-11-16 2003-05-30 3M Innovative Properties Company Methods and compositions related to irm compounds and toll-like receptor pathways
US20050070556A1 (en) 2001-11-27 2005-03-31 Anadys Pharmaceuticals, Inc. 3-B-D-ribofuranosylthiazolo [4,5-d] pyridimine nucleosides and uses thereof
US6924271B2 (en) 2001-11-27 2005-08-02 Anadys Pharmaceuticals, Inc. 3-β-D-ribofuranosylthiazolo[4-5-d]pyridimine nucleosides and uses thereof
WO2003049762A2 (en) 2001-12-12 2003-06-19 Chiron Srl. Immunisation against chlamydia trachomatis
US6924293B2 (en) 2001-12-21 2005-08-02 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6888000B2 (en) 2001-12-21 2005-05-03 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6677349B1 (en) 2001-12-21 2004-01-13 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
WO2003068811A2 (en) 2002-02-13 2003-08-21 Chiron Srl Cytotoxic t-cell epitopes from chlamydia
WO2003082272A1 (en) 2002-03-29 2003-10-09 Chiron Corporation Substituted benzazoles and use thereof as raf kinase inhibitors
WO2003093306A2 (en) 2002-05-02 2003-11-13 Chir0N Srl Nucleic acids and proteins from streptococcus groups a & b
WO2003097641A2 (en) 2002-05-21 2003-11-27 Novartis Ag 1h-imidazo[4,5-c] quinoline derivatives in the treatment of protein kinase dependent diseases
US6743920B2 (en) 2002-05-29 2004-06-01 3M Innovative Properties Company Process for imidazo[4,5-c]pyridin-4-amines
WO2004005473A2 (en) 2002-07-02 2004-01-15 Chiron Corporation Hcv fusion proteins with modified ns3 domains
WO2004018455A1 (en) 2002-08-23 2004-03-04 Chiron Corporation Pyrrole based inhibitors of glycogen synthase kinase 3
WO2004041157A2 (en) 2002-09-13 2004-05-21 Chiron Corporation Group b streptococcus vaccine
WO2004060308A2 (en) 2002-12-27 2004-07-22 Chiron Corporation Thiosemicarbazones as anti-virals and immunopotentiators
WO2004064759A2 (en) 2003-01-21 2004-08-05 Chiron Corporation Use of tryptanthrin compounds for immune potentiation
WO2004064715A2 (en) 2003-01-23 2004-08-05 M N L Pharma Limited Polyhydroxylated pyrrolizidine
WO2004071459A2 (en) 2003-02-13 2004-08-26 3M Innovative Properties Company Methods and compositions related to irm compounds and toll-like receptor 8
WO2004076677A2 (en) 2003-02-26 2004-09-10 Institute For Research In Biomedicine Monoclonal antibody production by ebv transformation of b cells
WO2004087153A2 (en) 2003-03-28 2004-10-14 Chiron Corporation Use of organic compounds for immunopotentiation
WO2004092360A2 (en) 2003-04-10 2004-10-28 Chiron Corporation The severe acute respiratory syndrome coronavirus
WO2005002619A2 (en) 2003-06-26 2005-01-13 Chiron Corporation Immunogenic compositions for chlamydia trachomatis
WO2005032582A2 (en) 2003-07-31 2005-04-14 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
WO2005025614A2 (en) 2003-09-15 2005-03-24 Glaxo Group Limited Improvements in vaccination

Non-Patent Citations (163)

* Cited by examiner, † Cited by third party
Title
"Current Status of Immunological Adjuvants", ANN. REV. IMMUNOL., vol. 4, 1986, pages 369 - 388
"Remington's Pharmaceutical Sciences", 1991, MACK PUB. CO.
"Vaccine Design: The Subunit and Adjuvant Approach", 1995, PLENUM PRESS, article "Chapter 10"
A. KYDONIEUS: "Treatise on Controlled Drug Delivery", 1992, MARCEL DEKKER, INC.
AHMAD, CHAPNICK, INFECT DIS CLIN NORTH AM, vol. 13, 1999, pages 113 - 133
ALLISON, BYARS, RES IMMUNOL, vol. 143, 1992, pages 519 - 25
ANDRIANOV ET AL.: "Preparation of hydrogel microspheres by coacervation of aqueous polyphophazene solutions", BIOMATERIALS, vol. 19, no. 1-3, 1998, pages 109 - 115
B.N. FIELDS AND D.M. KNIPE: "Fundamental Virology. 2nd Ed.", 1991
BARR ET AL.: "ISCOMs and other saponin based adjuvants", ADVANCED DRUG DELIVERY REVIEWS, vol. 32, 1998, pages 247 - 271
BEIGNON ET AL.: "The LTR72 Mutant of Heat-Labile Enterotoxin of Escherichia coli Enhances the Ability of Peptide Antigens to Elicit CD4+ T Cells and Secrete Gamma Interferon after Coapplication onto Bare Skin", INFECTION AND IMMUNITY, vol. 70, no. 6, 2002, pages 3012 - 3019
BELL, PEDIATR INFECT DIS J, vol. 19, 2000, pages 1187 - 1188
BHAGAT ET AL.: "CpG penta- and hexadeoxyribonucleotides as potent immunomodulatory agents", BBRC, vol. 300, 2003, pages 853 - 861
BIOCHIM BIOPHYS ACTA, vol. 1702, no. 2, 1 November 2004 (2004-11-01), pages 145
BIOCONJUGATE TECHNIQUES, 1996
BJUNE ET AL., LANCET, vol. 338, 1991
BLACKWELL ET AL.: "CpG-A-Induced Monocyte IFN-gamma-Inducible Protein-10 Production is Regulated by Plasmacytoid Dendritic Cell Derived IFN-alpha", J. IMMUNOL., vol. 170, no. 8, 2003, pages 4061 - 4068
BUTTERY, MOXON, J R COIL PHYSICIANS LONG, vol. 34, 2000, pages 163 - 168
CAN JBIOCHEM CELL BIOL., vol. 62, no. 5, May 1984 (1984-05-01), pages 270 - 5
CARINE DELEUZE-MASQUEFA, GREGORI GEREBTZOFF, GUY SUBRA, JEAN-ROCH FABREGUETTES, ANNABEL OVENS, MAELLE CARRAZ, MARIE-PAULE STRUB, J: "Design and synthesis of novel imidazo[1,2-a]quinoxalines as PDE4 inhibitor", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 12, no. 5, 2004, pages 1129 - 1139
CARINE DELEUZE-MASQUEFA: "Design and synthesis of novel imidazo[1,2-a]quinoxaolines as PDE4 inhibitors.", BIOORGANIC AND MEDICINAL CHEMISTRY, vol. 12, 2004, pages 1129 - 1139
CECCARELLI S ET AL: "Imidazo[1,2-a]quinoxalin-4-amines: A novel class of nonxanthine A1-adenosine receptor antagonists", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 33, no. 12, December 1998 (1998-12-01), pages 943 - 955, XP004160900, ISSN: 0223-5234 *
CHAUX P ET AL.: "Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes", J. EXP. MED., vol. 189, 1999, pages 767 - 78
CHUN-HE LIU, BO WANG, WEI-ZHANG LI, LIU-HONG YUN, YING LIU, RUI-BING SU, JIN LI, HE LIU: "Design, synthesis, and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine A1 receptor antagonist", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 12, no. 17, 2004, pages 4701 - 4707
CHUN-HE LIU: "Design, synthesis and biological evaluation of novel 4-alkylamino-1-hydroxymethylimidazo[1,2-a]quinoxalines as adenosine Al receptor antagonists.", BIOORGANIC AND MEDICINAL CHEMISTRY, vol. 12, 2004, pages 4701 - 4707
CONSTANTINO ET AL., VACCINE, vol. 10, 1992, pages 691 - 698
CONSTANTINO ET AL., VACCINE, vol. 17, 1999, pages 1251 - 1263
COOPER, PHARM BIOTECHNOL, vol. 6, 1995, pages 559 - 80
CRC, CHEMISTTY OFPROTEIN CONJUGATION AND CROSS-LINKING, 1993
CRUSE ET AL: "Conjugate Vaccines", vol. 10, pages: 48 - 114
DALE, INFECT DISCLIN NORTH AM, vol. 13, 1999, pages 227 - 43
DALE, VACCINE, vol. 14, no. 10, pages 944 - 948
DALE, VACCINE, vol. 17, 1999, pages 193 - 200
DAVIS, I.D. ET AL.: "Rational approaches to human cancer immunotherapy", JOURNAL OF LEUKOCYTE BIOLOGY, vol. 23, 2003, pages 3 - 29
DEL GUIDICE ET AL., MOLECULAR ASPECTS OF MEDICINE, vol. 19, 1998, pages 1 - 70
DELEUZE-MASQUEFA, CARINE ET AL: "Design and synthesis of novel imidazo[1,2-a]quinoxalines as PDE4 inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY , 12(5), 1129-1139 CODEN: BMECEP; ISSN: 0968-0896, 2004, XP002440985 *
DEREK T O'HAGAN, NICHOLAS M. VALIANTE, RECENT ADVANCES IN VACCINE ADJUVANTS AND DELIVERY SYSTEMS
DERMINE, S. ET AL.: "Cancer Vaccines and Immunotherapy", BRITISH MEDICAL BULLETIN, vol. 62, 2002, pages 149 - 162
DOMENIGHINI ET AL., MOL. MICROBIOL, vol. 15, no. 6, 1995, pages 1165 - 1167
DREENSEN, VACCINE, vol. 15, 1997, pages 2 - 6
E. NEGISHI.: "Oranopalladium Chemistry for Organic Synthesis", 2002, JOHN WILEY & SONS LTD.
EDWARD B. ROCHE: "Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
ESPINOZA-DELGADO I.: "Cancer Vaccines", THE ONCOLOGIST, vol. 7, no. 3, 2002, pages 20 - 33
EVANS ET AL., EXPERT REV VACCINES, vol. 2, 2003, pages 219 - 229
FERRETTI ET AL., PNAS USA, vol. 98, 2001, pages 4658 - 4663
FREY ET AL.: "Comparison of the safety, tolerability, and immunogenicity of a MF59-adjuvanted influenza vaccine and a non- adjuvanted influenza vaccine in non-elderly adults", VACCINE, vol. 21, 2003, pages 4234 - 4237
FUSKASAWA ET AL., VACCINE, vol. 17, 1999, pages 2951 - 2958
GASTOFSSON ET AL., N. ENGL. J. MED., vol. 334, 1996, pages 349 - 355
GAUDERNACK G: "T cell responses against mutant ras: a basis for novel cancer vaccines", IMMUNOTECHNOLOGY, vol. 2, 1996, pages 3 - 9
GEO KARMAS., PAUL E, SPOERRI: "Displacements and nuclear substitutions on hydroxypyrazines.", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 78, 1956, pages 4071 - 7
GERBER ET AL.: "Human Papillomavirus Virus-Like Particles Are Efficient Oral Immunogens when Coadministered with Escherichia coli Heat-Labile Entertoxin Mutant R192G or CpG", JOURNAL OF VIROLOGY, vol. 75, no. 10, 2001, pages 4752 - 4760
GERLICH ET AL., VACCINE, vol. 8, 1990, pages 63 - 68,79-80
GLUCK ET AL.: "New Technology Platforms in the Development of Vaccines for the Future", VACCINE, vol. 20, 2002, pages B10 - B16
GOLD P ET AL.: "Specific carcinoembryonic antigens of the human digestive system", J. EXP. MED., vol. 122, 1965, pages 467 - 8
GOLDBLATT, J. MED. MICROBIOL., vol. 47, 1998, pages 663 - 567
GREENE, T.W.: "Protective Groups in Organic Synthesis. 1st Ed.", 1981, JOHN WILEY & SONS
HAN ET AL.: "Impact of Vitamin E on Immune Function and Infectious Diseases in the Aged at Nutrition", IMMUNE FUNCTIONS AND HEALTH EUROCONFERENCE, 9 June 2005 (2005-06-09)
HARIHARAN ET AL., CANCER RES, vol. 55, 1995, pages 3486 - 9
HERMANSON, BIOCONJUGATE TECHNIQUES, 1996
HOUGHTON ET AL., HEPATOLOGY, vol. 14, 1991, pages 381
HSU ET AL., CLIN LIVER DIS, vol. 3, 1999, pages 901 - 915
INFECT IMMUN, vol. 69, no. 5, May 2001 (2001-05-01), pages 3323 - 3334
INFECT IMMUN., vol. 65, no. 11, November 1997 (1997-11-01), pages 4476 - 4482
INFECT IMMUN., vol. 67, no. 10, October 1999 (1999-10-01), pages 5395
INFECT IMMUN., vol. 72, no. 10, October 2004 (2004-10-01), pages 6148
INFECT IMMUN., vol. 72, no. 7, July 2004 (2004-07-01), pages 3829
INFECT IMMUN., vol. 9, no. 5, 6 May 2001 (2001-05-06), pages 3510 - 3515
INFECT LMMUN, vol. 1, no. 11, 7 January 2003 (2003-01-07), pages 374 - 383
INFECT LMMUN., vol. 71, no. 10, October 2003 (2003-10-01), pages 5498 - 504
IWARSON, APMIS, vol. 103, 1995, pages 321 - 326
J AUTOIMMUN., vol. 2, June 1989 (1989-06-01), pages 81
J CLIN MICROBIOL, vol. 37, no. 12, December 1999 (1999-12-01), pages 3997
JEDRZEJAS, MICROBIOL MOL BIOL REV, vol. 65, 2001, pages 187 - 207
JIRO ADACHI, NOBUHIRO SATO: "Pyrazines. 1. Syntheses of 2,3-dihydroxypyrazines and their derivatives.", JOURNAL OF ORGANIC CHEMISTRY, vol. 37, no. 2, 1972, pages 221 - 5
JIRO TSUJI: "Palladium Reagents and Catalysts: Innovations in Organic Chemistry", JOHN WILEY & SONS LTD.
JOHNSON ET AL., BIOORG MED CHEM LETT, vol. 9, 1999, pages 2273 - 2278
JONES: "Resiquimod 3M", CURR OPIN INVESTIG DRUGS, vol. 4, no. 2, 2003, pages 214 - 218
KALMAN ET AL., NATURE GENETICS, vol. 21, 1999, pages 385 - 389
KANDIMALLA ET AL.: "Divergent synthetic nucleotide motif recognition pattern: design and development of potent immunomodulatory oligodeoxyribonucleotide agents with distinct cytokine induction profiles", NUCLEIC ACIDS RESEARCH, vol. 31, no. 9, 2003, pages 2393 - 2400
KANDIMALLA ET AL.: "Secondary structures in CpG oligonucleotides affect immunostimulatory activity", BBRC, vol. 306, 2003, pages 948 - 953
KANDIMALLA ET AL.: "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic CpG DNAs", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 31, 2003, pages 654 - 658
KANDIMALLA ET AL.: "Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic GpG DNAs", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 31, 2003, pages 664 - 658
KRIEG: "CpG motifs: the active ingredient in bacterial extracts?", NATURE MEDICINE, vol. 9, no. 7, 2003, pages 831 - 835
KRIEG: "From A to Z on CpG", TRENDS IN IMMUNOLOGY, vol. 23, no. 2, 2002, pages 64 - 65
KURODA ET AL., LANCET, vol. 357, no. 9264, 2001, pages 1225 - 1240,1218-1219
LEE, DIFFUSION-CONTROLLED MATRIX SYSTEMS, pages 155 - 198
LENZ ET AL.: "Papillomarivurs-Like Particles Induce Acute Activation of Dendritic Cells", JOURNAL OF IMMUNOLOGY, 2001, pages 5246 - 5355
LINDBERG, VACCINE, vol. 17, no. 2, 1999, pages 28 - 36
LIVINGSTON PO ET AL.: "Carbohydrate vaccines that induce antibodies against cancer: Previous experience and future plans", CANCER IMMUNOL. IMMUNOTHER., vol. 45, 1997, pages 10 - 9
LIVINGSTON PO ET AL.: "Carbohydrate vaccines that induce antibodies against cancer: Rationale", CANCER IMMUNOL. IMMUNOTHER., vol. 45, 1997, pages 1 - 6
MACKAY, B., POCKET PETS, ANIMAL ISSUES, vol. 32, no. 1, 2001
MATTSON, CHAN, SCIENCE, vol. 301, no. 1 1, 2003, pages 847 - 9
MCCLUSKIE ET AL.: "Parenteral and mucosal prime-boost immunization strategies in mice with hepatitis B surface antigen and CpG DNA", FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY, vol. 32, 2002, pages 179 - 185
MCMICHAEL, VACCINE, vol. L9, no. 1, 2000, pages 101 - 107
MERALDI ET AL.: "OM-174, a New Adjuvant with a Potential for Human Use, Induces a Protective Response with Administered with the Synthetic C-Terminal Fragment 242-310 from the circumsporozoite protein of Plasmodium berghei", VACCINE, vol. 21, 2003, pages 2485 - 2491
MISCHLER, METCALFE, VACCINE, vol. 20, no. 5, 2002, pages B17 - 23
MISHELL AND SHIIGI: "Selected Methods in Cellular Immunology", 1980, W.H. FREEMAN AND CO
MMWR MORB MORTAL WKLY REP, vol. 47, no. 1, 16 January 1998 (1998-01-16), pages 12,9
MOINGEON P: "Cancer vaccines", VACCINE, vol. 19, 2001, pages 1305 - 1326
MURRAY ET AL.: "Medical Microbiology", 2002
N. SATO.: "Product class 14: pyrazines.", SCIENCE OF SYNTHESIS, vol. 16, 2004, pages 751 - 844
NIIKURA ET AL.: "Chimeric Recombinant Hepatitis E Virus-Like Particles as an Oral Vaccine Vehicle Presenting Foreign Epitopes", VIROLOGY, vol. 293, 2002, pages 273 - 280
OFFRINGA R ET AL.: "Design and evaluation of antigen-specific vaccination strategies against cancer", CURRENT OPIN. IMMUNOL., vol. 2, 2000, pages 576 - 582
O'HAGAN: "Vaccine Adjuvants: Preparation Methods and Research Protocols", vol. 42, article "Methods in Molecular Medicine series"
OLD LJ ET AL.: "New paths in human cancer serology", J. EXP. MED., vol. 187, 1998, pages 1163 - 7
OTT ET AL.: "Vaccine Design: The Subunit and Adjuvant Approach", 1995, PLENUM PRESS, article "MF59 -- Design and Evaluation of a Safe and Potent Adjuvant for Human Vaccines", pages: 277 - 296
PAJAK ET AL.: "The Adjuvant OM-174 induces both the migration and maturation of murine dendritic cells in vivo", VACCINE, vol. 21, 2003, pages 836 - 842
PARTIDOS ET AL.: "Heat-labile enterotoxin of Escherichia coli and its site-directed mutant LTK63 enhance the proliferative and cytotoxic T-cell responses to intranasally co-immunized synthetic peptides", IMMUNOL. LETT., vol. 67, no. 3, 1999, pages 209 - 216
PAYNE ET AL.: "Protein Release from Polyphosphazene Matrices", ADV. DRUG. DELIVERY REVIEW, vol. 31, no. 3, 1998, pages 185 - 196
PEPPOLONI ET AL.: "Mutants of the Escherichia coli heat-labile enterotoxin as safe and strong adjuvants for intranasal delivery of vaccines", VACCINES, vol. 2, no. 2, 2003, pages 285 - 293
PHARM RES., vol. 21, no. 12, December 2004 (2004-12-01), pages 2148 - 52
PINE ET AL.: "Intranasal immunization with influenza vaccine and a detoxified mutant of heat labile enterotoxin from Escherichia coli (LTK63", J. CONTROL RELEASE, vol. 85, no. 1-3, 2002, pages 263 - 270
PINTO ET AL.: "Cellular Immune Responses to Human Papillomavirus (HPV)-16 L Healthy Volunteers Immunized with Recombinant HPV-16 L Virus-Like Particles", JOURNAL OF INFECTIOUS DISEASES, vol. 188, 2003, pages 327 - 338
PIZZA ET AL., SCIENCE, vol. 287, 2000, pages 1816 - 1820
PIZZA ET AL.: "LTK63 and LTR72, two mucosal adjuvants ready for clinical trials", INT. J. MED. MICROBIOL, vol. 290, no. 4-5, 2000, pages 455 - 461
PIZZA ET AL.: "Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants", VACCINE, vol. 19, 2001, pages 2534 - 2541
PLANTE ET AL., J INFECTIOUS DISEASE, vol. 182, 2000, pages 848 - 855
PLOTKIN AND ORENSTEIN: "Vaccines. 4th Ed.", 2004
PODDA, DEL GIUDICE, EXPERT REV VACCINES, vol. 2, 2003, pages 197 - 203
PODDA, VACCINE, vol. 19, 2001, pages 2673 - 2680
PODDA: "The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine", VACCINE, vol. 19, 2001, pages 2673 - 2680
POPE ET AL., CELLULAR IMMUNOLOGY, vol. 162, 1995, pages 333 - 339
POWELL & NEWMAN: "Vaccine Design...", 1995, PLENUM
PRESCOTT: "Methods in Cell Biology", vol. XIV, 1976, ACADEMIC PRESS, pages: 33
PRICE ET AL., INFECTION AND IMMUNITY, vol. 71, no. 1, 2004, pages 277 - 283
PROC NATL ACAD SCI US A, vol. 101, no. 34, 24 August 2004 (2004-08-24), pages 12652
RAMSAY ET AL., LANCET, vol. 357, no. 9251, 2001, pages 195 - 196
RAPPUOLI ET AL., TIBTECH, vol. 9, 1991, pages 232 - 238
RAZ ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 9519 - 9523
READ ET AL., NUCLEIC ACIDS RES, vol. 28, 2000, pages 1397 - 406
RON, LANGER, ERODIBLE SYSTEMS, pages 199 - 224
ROSENBERG SA: "A new era for cancer immunotherapy based on the genes that encode cancer antigens", IMMUNITY, vol. 10, 1999, pages 281 - 7
ROSENBERG SA: "Cancer vaccines based on the identification of genes encoding cancer regression antigens", IMMUNOL. TODAY, vol. 18, 1997, pages 175 - 82
ROSENBERG SA: "Progress in human tumor immunology and immunotherapy", NATURE, vol. 411, 2001, pages 380 - 384
ROSENQIST ET AL., DEV. BIOL. STRAND, vol. 92, 1998, pages 323 - 333
ROSS ET AL., VACCINE, vol. 19, 2001, pages 135 - 142
RUBIN, PEDIATR CLIN NORTH AM, vol. 47, 2000, pages 269 - 285
RYAN ET AL.: "Mutants of Escherichia coli Heat-Labile Toxin Act as Effective Mucosal Adjuvants for Nasal Delivery of an Acellular Pertussis Vaccine: Differential Effects of the Nontoxic AB Complex and Enzyme Activity on Thl and Th2 Cells", INFECTION AND IMMUNITY, vol. 67, no. 12, 1999, pages 6270 - 6280
SABROE ET AL., JL, 2003, pages 1630 - 5
SAHIN U ET AL.: "Serological identification of human tumor antigens", CURR. OPIN. IMMUNOL., vol. 9, 1997, pages 709 - 16
SCHARTON-KERSTEN ET AL.: "Transcutaneous Immunization with Bacterial ADP-Ribosylating Exotoxins, Subunits and Unrelated Adjuvants", INFECTION AND IMMUNITY, vol. 68, no. 9, 2000, pages 5306 - 5313
SCHUCHAT, LANCER, vol. 353, no. 9146, 1999, pages 51 - 6
SHIRAI ET AL., J. INFECT. DIS, vol. 181, no. 3, 2000, pages S524 - S527
SIGNORELLI, HADDEN, INT IMMUNOPHARMACOL, vol. 3, no. 8, 2003, pages 1177 - 86
SINGH ET AL., J. CONT. RELE., vol. 70, 2001, pages 267 - 276
SJOLANDER ET AL.: "Uptake and adjuvant activity of orally delivered saponin and ISCOM vaccines", ADVANCED DRUG DELIVERY REVIEWS, vol. 32, 1998, pages 321 - 338
STANLEY: "Imiquimod and the imidazoquinolines: mechanism of action and therapeutic potential", CLIN EXP DERMATOL, vol. 27, no. 7, 2002, pages 571 - 577
STEPHANIE PARRA: "Imidazo[1,2-a]quinoxalines: synthesis and cyclic nucleotide phosphodiesterase inhibitory activity.", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 36, 2001, pages 255 - 264
SUTTER ET AL., PEDIATR CLIN NORTH AM, vol. 47, 2000, pages 287 - 308
T. HIGUCHI, V. STELLA: "Pro-drugs as Novel Delivery Systems", vol. 14, A.C.S. SYMPOSIUM SERIES
TAYLOR-PAPADIMITRIOU J: "Biology, biochemistry and immunology of carcinoma- associated mucins", IMMUNOL. TODAY, vol. 18, 1997, pages 105 - 7
TETTELIN ET AL., SCIENCE, vol. 287, 2000, pages 1809 - 1815
THEOBALD M ET AL.: "Targeting p53 as a general tumor antigen", PROC. NATL. ACAD. SCI. USA, vol. 92, 1995, pages 11993 - 7
THOMPSON ET AL., J LEUKOC BIOL, 2005, pages 78
THOMPSON ET AL., METHODS IN MOLECULAR MEDICINE, vol. 94, 2003, pages 255 - 266
VAN DEN EYNDE B ET AL.: "New tumor antigens recognized by T cells", CURR. OPIN. IMMUNOL., vol. 7, 1995, pages 674 - 81
VASILAKOS ET AL., CELL IMMUNOL., vol. 204, no. 1, 2000, pages 64 - 74
W.K. JOKLIK: "Virology. 3rd Ed.", 1988
WATSON, PEDIATR INFECT DIS J, vol. 19, 2000, pages 331 - 332
WONG ET AL., J CLIN PHARMACOL, vol. 43, no. 7, 2003, pages 735 - 42
WU ET AL., ANTIVIRAL RES., vol. 64, no. 2, 2004, pages 79 - 83
ZHAO X-J ET AL.: "GD2 oligosaccharide: target for cytotoxic T lymphocytes", J. EXP. MED., vol. 182, 1995, pages 67 - 74
ZHU ET AL., VACCINE, vol. 22, 2004, pages 660 - 669
ZIMMERMAN, SPANN, AM FAN PHYSICIAN, vol. 59, 1999, pages 113 - 118,125-126

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190104519A (en) * 2016-11-09 2019-09-10 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Methods and Compositions for Acquired Immune Regulation
CN110234401A (en) * 2016-11-09 2019-09-13 德克萨斯大学系统董事会 The method and composition adjusted for adaptive immunity
EP3538218A4 (en) * 2016-11-09 2020-06-17 The Board of Regents of The University of Texas System Methods and compositions for adaptive immune modulation
AU2017356673B2 (en) * 2016-11-09 2023-11-09 Pulmotect, Inc. Methods and compositions for adaptive immune modulation
CN110234401B (en) * 2016-11-09 2024-03-01 德克萨斯大学系统董事会 Methods and compositions for adaptive immunomodulation

Also Published As

Publication number Publication date
US8173657B2 (en) 2012-05-08
EP2010537A1 (en) 2009-01-07
US20130101622A1 (en) 2013-04-25
WO2007109813A1 (en) 2007-09-27
ES2376492T3 (en) 2012-03-14
EP2357184B1 (en) 2015-02-25
ES2536426T3 (en) 2015-05-25
EP2010537B1 (en) 2011-12-28
CA2646539A1 (en) 2007-09-27
ATE539079T1 (en) 2012-01-15
US20090311288A1 (en) 2009-12-17

Similar Documents

Publication Publication Date Title
EP2010537B1 (en) Imidazoquinoxaline compounds as immunomodulators
EP2007765B1 (en) Immunopotentiating compounds
RU2415857C2 (en) Imidazoquinoline compounds
US20100010217A1 (en) Methods for the preparation of imidazole-containing compounds
KR101853513B1 (en) Compounds (cystein based lipopeptides) and compositions as tlr2 agonists used for treating infections, inflammations, respiratory diseases etc.
JP5988492B2 (en) Immunogenic composition comprising a TLR activity modulator
EP2473508B1 (en) Compounds and compositions as tlr activity modulators
US20100226931A1 (en) Compounds for immunopotentiation
WO2006002422A9 (en) Compounds for immunopotentiation
TW200951132A (en) Compounds and compositions as TLR activity modulators
WO2011057148A1 (en) Compounds and compositions as tlr-7 activity modulators
AU2012209025A1 (en) Imidazoquinoline compounds

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 2010537

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

17P Request for examination filed

Effective date: 20120217

17Q First examination report despatched

Effective date: 20120504

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 37/00 20060101ALI20140402BHEP

Ipc: C07H 7/06 20060101ALI20140402BHEP

Ipc: A61K 31/4985 20060101ALI20140402BHEP

Ipc: C07D 487/04 20060101AFI20140402BHEP

INTG Intention to grant announced

Effective date: 20140425

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AC Divisional application: reference to earlier application

Ref document number: 2010537

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007040428

Country of ref document: DE

Effective date: 20150409

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 711887

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150415

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20150227

Year of fee payment: 9

Ref country code: DE

Payment date: 20150317

Year of fee payment: 9

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2536426

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20150525

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20150318

Year of fee payment: 9

Ref country code: FR

Payment date: 20150309

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20150311

Year of fee payment: 9

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20150225

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 711887

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150225

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150625

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150526

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007040428

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150331

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150331

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150323

26N No opposition filed

Effective date: 20151126

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602007040428

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20160323

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20161130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160331

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160323

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20161001

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20070323

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150625

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150323

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160324

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20181205